651
|
Colleoni A, Galli G, Dallanoce C, De Amici M, Gorostiza P, Matera C. Light-Activated Pharmacological Tools for Exploring the Cholinergic System. Med Res Rev 2025. [PMID: 40123150 DOI: 10.1002/med.22108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
Cholinergic transmission plays a critical role in both the central and peripheral nervous systems, affecting processes such as learning, memory, and inflammation. Conventional cholinergic drugs generally suffer from poor selectivity and temporal precision, leading to undesired effects and limited therapeutic efficacy. Photopharmacology aims to overcome the limitations of traditional drugs using photocleavable or photoswitchable ligands and spatiotemporal patterns of illumination. Spanning from muscarinic and nicotinic modulators to cholinesterase inhibitors, this review explores the development and application of light-activated compounds as tools for unraveling the role of cholinergic signaling in both physiological and pathological contexts, while also paving the way for innovative phototherapeutic approaches.
Collapse
Grants
- This research was supported by the European Union-Next Generation EU, Mission 4, Component 1 (CUP J53C24002040004), EU Horizon 2020 Framework Programme for Research and Innovation, European Innovation Council Pathfinder (PHOTOTHERAPORT, 101130883), Human Brain Project (WaveScalES, SGA3, 945539), Information and Communication Technologies (Deeper, ICT-36-2020-101016787), and Piano di Sostegno alla Ricerca 2023 (Azione A, Linea 2, PSR2023_DIP_021_CMATE). It was also supported by the Government of Catalonia (CERCA Programme; AGAUR 2021-SGR-01410), Spanish Ministry of Science and Innovation (DEEP RED, grant PID2019-111493RB-I00; EPILLUM, grant AEI/10.13039/501100011033; and Research Network in Biomedicine eBrains-Spain, RED2022-134823-E).
Collapse
Affiliation(s)
- Alessio Colleoni
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Giulia Galli
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Clelia Dallanoce
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Marco De Amici
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Pau Gorostiza
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology, Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Carlo Matera
- Section of Medicinal Chemistry "Pietro Pratesi", Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
652
|
Hafez HM, Said BAM, Sayed AM, Alatwi E, Youssif BGM, Bräse S, El-Sherief HAM. Design, synthesis, antiproliferative activity, and molecular dynamics simulation of pyrazoline-based derivatives as dual EGFR and HER-2 inhibitors. RSC Adv 2025; 15:9265-9279. [PMID: 40161522 PMCID: PMC11951252 DOI: 10.1039/d5ra01169h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
The dual targeting of EGFR and HER2 is an established anticancer strategy. A novel series including two distinct scaffolds, A (chalcone-based compounds, 4a-n) and B (pyrazoline-based compounds, 5a-n), was developed and synthesized. The antiproliferative efficacy of 4a-n and 5a-n was examined against a panel of four cancer cell lines. The findings indicated that pyrazoline derivatives 5a-n exhibited more efficacy than chalcone compounds 4a-n. Compounds 4n, 5d, and 5g were identified as the most effective antiproliferative derivatives. These compounds were further investigated as dual EGFR/Her2 inhibitors. Compound 5d inhibited EGFR-TK and HER2 significantly, with IC50 values of 0.126 and 0.061 μM, respectively. Moreover, compound 5d can induce a percentage of pre-G1 apoptosis by 78.53% in cell cycle analysis and cause early apoptosis with necrosis percent of 5.28. Docking and MD simulation illustrated the significant cytotoxic activity of the 5d compound and how it can be a promising scaffold with anticancer activity.
Collapse
Affiliation(s)
- Hani Mohamed Hafez
- Pharmaceutical Chemistry Branch, College of Pharmacy, Al-Esraa University College Baghdad Iraq
| | | | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University 62513 Beni Suef Egypt
- Department of Pharmacognosy, Collage of Pharmacy, Almaaqal University 61014 Basrah Iraq
| | - Eid Alatwi
- Department of Pharmacology, College of Pharmacy, Jouf University Sakaka 72341 Aljouf Saudi Arabia
| | - Bahaa G M Youssif
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt +20-1098294419
| | - Stefan Bräse
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology 76131 Karlsruhe Germany
| | - Hany A M El-Sherief
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Deraya University Minia Egypt
| |
Collapse
|
653
|
Llewellyn A, Barrow-McGee R, Stevenson J, Gore J, Naidoo K. Protocol for perfusing human axillary lymph nodes ex vivo to study structure and function in real time. STAR Protoc 2025; 6:103624. [PMID: 39913290 PMCID: PMC11848449 DOI: 10.1016/j.xpro.2025.103624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/01/2024] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
Lymph nodes regulate immunity and maintain fluid balance in health and disease. Here, we present a protocol that uses normothermic perfusion to sustain patient-derived lymph nodes ex vivo for up to 24 h to study their structure and function. We describe steps for setting up both thermoregulatory and perfusion circuits, cannulating human lymph nodes, and perfusion. This protocol can be used to study how human lymph nodes change in cancer and other diseases, and/or in response to perturbations, including drugs. For complete details on the use and execution of this protocol, please refer to Barrow-McGee et al.1.
Collapse
Affiliation(s)
- Amy Llewellyn
- Translational Pathology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Rachel Barrow-McGee
- Toby Robins Breast Cancer Now Research Centre, Breast Cancer Research Division, The Institute of Cancer Research, London, UK
| | - Julia Stevenson
- King's Health Partners Cancer Biobank, Guy's Comprehensive Cancer Centre, London, UK
| | - Jasmine Gore
- Translational Pathology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Kalnisha Naidoo
- Translational Pathology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK; Department of Cellular Pathology, King's College Hospital, London, UK.
| |
Collapse
|
654
|
Nie P, Wu Y, Robinson J, Mekala S, Lee VMY, Li YM. In Situ Labeling of Pathogenic Tau Using Photo-Affinity Chemical Probes. ACS Chem Biol 2025; 20:581-591. [PMID: 40079621 DOI: 10.1021/acschembio.5c00073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Tau aggregation plays a crucial role in the development of Alzheimer's disease (AD). Developing specific techniques that can isolate pathogenic tau from brain tissue is important for understanding tauopathies and advancing targeted therapies. Here, we develop photoaffinity small molecular probes and a novel method for in situ tissue labeling and investigate their activity in interacting with tau in cells and AD patient brains. Based on the reported chemical structures of tau PET tracers, we designed and synthesized two tau-specific probes, namely, Tau-2 and Tau-4. After validation in cell, mouse model, and patient brain samples, our photolabeling results suggested that Tau-2 effectively labels soluble tau in cell and mouse models, while Tau-4 selectively binds high-molecular-weight tau aggregates in late-stage AD patient brain tissues. Proteomic analysis verified the specific isolation of pathogenic tau from AD brain samples. Collectively, these findings underscore the potential of our photoaffinity probes as powerful tools for investigating tau proteins and neurofibrillary tangles in neurodegenerative diseases.
Collapse
Affiliation(s)
- Pengju Nie
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - You Wu
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - John Robinson
- Department of Pathology and Laboratory Medicine, Institute on Aging, and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Shekar Mekala
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - Virginia M Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging, and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| |
Collapse
|
655
|
Momoli C, Arcadi A, Chiarini M, Morlacci V, Palombi L. Expanding Diversity of Fused Steroid-Quinoline Hybrids by Sequential Amination/Annulation/Aromatization Reactions. J Org Chem 2025; 90:3951-3963. [PMID: 40052426 PMCID: PMC11934142 DOI: 10.1021/acs.joc.4c02981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/01/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Viable alternative approaches to a variety of ring A and ring D-fused steroid-quinoline hybrids, along with ring A, D-fused, and/or ring A-fused, side chain-substituted steroid-bis-quinolines were explored by means of sequential amination/annulation/aromatization reactions of suitable ketosteroids with 2-acyl-substituted anilines. Key factors directing the chemoselective behavior of polyfunctionalized substrates were investigated. Remarkably, the use of TMSOTf as an alternative promoter/catalyst enabled the direct synthesis of the desired hybrids, avoiding the protection/deprotection steps of the conventional procedures when the starting substrates contained labile functional groups.
Collapse
Affiliation(s)
- Caterina Momoli
- Dipartimento
di Scienze Fisiche e Chimiche, Università
degli studi dell’Aquila, Via Vetoio, Coppito (AQ), 67100 L’Aquila,Italy
| | - Antonio Arcadi
- Dipartimento
di Scienze Fisiche e Chimiche, Università
degli studi dell’Aquila, Via Vetoio, Coppito (AQ), 67100 L’Aquila,Italy
| | - Marco Chiarini
- Dipartimento
di Bioscienze e Tecnologie Agroalimentari e Ambientali, Università degli studi di Teramo, Via R. Balzarini, 64110 Teramo, Italy
| | - Valerio Morlacci
- Dipartimento
di Scienze Fisiche e Chimiche, Università
degli studi dell’Aquila, Via Vetoio, Coppito (AQ), 67100 L’Aquila,Italy
| | - Laura Palombi
- Dipartimento
di Scienze Fisiche e Chimiche, Università
degli studi dell’Aquila, Via Vetoio, Coppito (AQ), 67100 L’Aquila,Italy
| |
Collapse
|
656
|
Hagras M, Ezzat HG, Abuelkhir AA, Mayhoub AS. Repurposing the phenylthiazole scaffold with 1,3,4-oxadiazole for selective, potent and well-tolerated antifungal activity. RSC Adv 2025; 15:9305-9319. [PMID: 40151538 PMCID: PMC11948216 DOI: 10.1039/d5ra00499c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Invasive fungal infections (IFIs) represent a critical health threat, particularly among immunocompromised individuals, with mortality rates reaching up to 50%. The growing resistance to existing antifungal therapies necessitates the development of novel agents. Here, we rationally designed phenylthiazole-based oxadiazole derivatives to enhance selectivity and potency against resistant fungal strains. Among the tested compounds, compound 35 (which emerged as a lead candidate) demonstrated potent activity against Candida albicans (MIC = 1-2 μg mL-1), Candida glabrata (MIC = 0.5-1 μg mL-1), and multidrug-resistant Candida auris (MIC = 2-4 μg mL-1), outperforming fluconazole and matching amphotericin B. Additionally, compound 35 showed minimal cytotoxicity (88% cell viability at 16 μg mL-1) and negligible hemolytic activity, indicating a superior safety profile.
Collapse
Affiliation(s)
- Mohamed Hagras
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University Cairo 11884 Egypt
| | - Hany G Ezzat
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University Cairo 11884 Egypt
| | - Abdelrahman A Abuelkhir
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University Cairo 11884 Egypt
| | - Abdelrahman S Mayhoub
- Department of Pharmaceutical Organic Chemistry, College of Pharmacy, Al-Azhar University Cairo 11884 Egypt
- Nanoscience Program, University of Science and Technology, Zewail City of Science and Technology October Gardens 6th of October Giza 12578 Egypt
| |
Collapse
|
657
|
Bandyopadhyay A, Sarkar R. Site-selective cleavage of peptides and proteins targeting aromatic amino acid residues. RSC Adv 2025; 15:9159-9179. [PMID: 40134686 PMCID: PMC11934106 DOI: 10.1039/d4ra08956a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
The site-selective cleavage of peptides and proteins at specific amino acid residues is an important strategy for the modification of biomolecules as it can potentially transmute the reactivity profile of the whole molecule. Moreover, precise cleavage of a specific amide bond in peptides and proteins has enormous applications in the domains of chemical biology, genetics, and protein engineering. Among the 20 proteinogenic amino acids, tryptophan (Trp, W), tyrosine (Tyr, Y), phenylalanine (Phe, F) and histidine (His, H) are classified as aromatic amino acids that maintain the function of protein folding through hydrophobic and π-π interactions. Thus, scissoring at a specific site of an aromatic amino acid may alter the structure and function of a peptide or protein. In the last 60-70 years, great success has been achieved in the development of methods for the aromatic amino acid (AAA)-selective cleavage of peptides and proteins. Generally, aromatic side chains are derivatized in the presence of specific reagents. Consequently, either the downstream or the upstream amide bond of the aromatic side chain is activated, and hydrolysis of the amide bond splits the peptide. Unfortunately, a systematic review covering this methodological development of the AAA-selective fission of peptide is lacking to date. Thus, in this review, we aim to showcase the up-to-date progress in the site-selective rupture of peptide bonds at aromatic amino acid residues with an emphasis on the postulated mechanisms, enabling future researchers to further drive progress in this research field.
Collapse
Affiliation(s)
- Ayan Bandyopadhyay
- Department of Chemistry, Chapra Government College Nadia-741123 West Bengal India
- Department of Higher Education, Government of West Bengal India
| | - Rajib Sarkar
- Department of Higher Education, Government of West Bengal India
- Department of Chemistry, Muragachha Government College Nadia-741154 West Bengal India
| |
Collapse
|
658
|
Bando Y, Klapper M, Herbst R, Sachse A, Stallforth P, Arndt HD. Semiautomated Total Synthesis of the Cyclic Lipodepsipeptide Anikasin. Org Lett 2025; 27:2559-2563. [PMID: 40062948 DOI: 10.1021/acs.orglett.5c00111] [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: 03/22/2025]
Abstract
The total synthesis of Pseudomonas-derived cyclic lipodepsipeptide anikasin was achieved. Using a depsipeptide building block and balanced protecting groups on the branching d-allo-Thr residue, the synthesis was established semiautomatically on a synthesizer. Buffered deprotections minimized side reactions and afforded synthetic anikasin and its enantiomer. Biological activity studies indicated that anikasin's mode of action is directly resulting from its physicochemical properties.
Collapse
Affiliation(s)
- Yuko Bando
- Friedrich Schiller University Jena, Institute for Organic Chemistry and Macromol. Chemistry, Humboldtstr. 10, D-07743 Jena, Germany
| | - Martin Klapper
- Leibniz Institute for Natural Product Research and Infection Biology─Hans Knöll Institute, Beutenbergstr. 11a, D-07745 Jena, Germany
| | - Rosa Herbst
- Leibniz Institute for Natural Product Research and Infection Biology─Hans Knöll Institute, Beutenbergstr. 11a, D-07745 Jena, Germany
| | - Anna Sachse
- Friedrich Schiller University Jena, Institute for Organic Chemistry and Macromol. Chemistry, Humboldtstr. 10, D-07743 Jena, Germany
| | - Pierre Stallforth
- Friedrich Schiller University Jena, Institute for Organic Chemistry and Macromol. Chemistry, Humboldtstr. 10, D-07743 Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology─Hans Knöll Institute, Beutenbergstr. 11a, D-07745 Jena, Germany
- Department of Paleobiotechnology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 07745 Jena, Germany
| | - Hans-Dieter Arndt
- Friedrich Schiller University Jena, Institute for Organic Chemistry and Macromol. Chemistry, Humboldtstr. 10, D-07743 Jena, Germany
| |
Collapse
|
659
|
Marimuthu SCV, Thangamariappan E, Kunjiappan S, Pandian SRK, Sundar K. New insights into iron uptake in Streptococcus mutans: evidence for a role of siderophore-like molecules. Arch Microbiol 2025; 207:96. [PMID: 40111578 DOI: 10.1007/s00203-025-04284-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/11/2025] [Accepted: 02/23/2025] [Indexed: 03/22/2025]
Abstract
Streptococcus mutans, a gram-positive coccus commonly found in the human oral cavity, is the primary causative agent of dental caries as well as infective endocarditis. Bacteria produce potent iron chelators called siderophores to absorb iron. Because, there are few studies on siderophore-mediated iron transport in S. mutans, the current study investigates the presence of such a mechanism in S. mutans GS-5. Deferration of culture medium and different concentrations of 2, 2'-Bipyridyl has been used to simulate iron-restricted conditions. Iron restriction alters the colony morphology and slows bacterial growth. Cross-feeding conditioned medium into an iron-restricted medium promotes bacterial growth, indicating the presence of siderophore-like molecules. This was further confirmed by Chrome Azurol S (CAS) assay and Modified CAS-agar assay. Cśaky's and Arnow's assays detected the presence of hydroxamate and catecholate-type molecules in optimal and iron-restricted conditions, respectively. Further, the siderophore-like molecules were extracted and purified with thin layer chromatography (TLC). TLC elutes were also found to be positive for iron-chelation in CAS-agar assay and aided growth of S. mutans under iron-restricted conditions. LC-MS analysis of culture supernatants under iron-restricted conditions identified iron-binding small molecules, including a catechol structural motif. Computational analysis utilizing KEGG and BLASTp suggested homologues of siderophore biosynthesis and transport proteins, including genes associated with mutanobactin production. These findings indicate a possible siderophore-mediated iron uptake mechanism in S. mutans GS-5, warranting further molecular studies and advanced spectroscopic characterization of this unidentified siderophore. Once confirmed, this mechanism can be used as a potential drug target to control streptococcal infection.
Collapse
Affiliation(s)
- Shakti Chandra Vadhana Marimuthu
- Department of Biotechnology, School of Bio, Chemical and Processing Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, 626126, India
| | - Esakkimuthu Thangamariappan
- Department of Biotechnology, School of Bio, Chemical and Processing Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, 626126, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, School of Bio, Chemical and Processing Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, 626126, India
| | - Sureshbabu Ram Kumar Pandian
- Department of Biotechnology, School of Bio, Chemical and Processing Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, 626126, India
| | - Krishnan Sundar
- Department of Biotechnology, School of Bio, Chemical and Processing Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, 626126, India.
| |
Collapse
|
660
|
Hermawan A, Hanif N, Putri DDP, Fatimah N, Prasetio HH. Citrus flavonoids for overcoming breast cancer resistance to methotrexate: identification of potential targets of nobiletin and sinensetin. Discov Oncol 2025; 16:365. [PMID: 40111633 PMCID: PMC11926326 DOI: 10.1007/s12672-025-02116-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Breast cancer is a potentially fatal illness that affects millions of women worldwide. Methotrexate (MTX) may be beneficial for treating breast cancer; however, high doses and prolonged use can cause drug resistance. Although certain citrus flavonoids-nobiletin, sinensetin, tangeretin, hesperidin, hesperetin, and naringenin-may overcome resistance to chemotherapy, no study has investigated MTX resistance. This study investigated the potential of natural chemicals, specifically nobiletin and sinensetin, to overcome MTX resistance in breast cancer cells using MTX-resistant MCF-7 (MCF-7/MTX) and MCF-7 cells. Protein targets of citrus flavonoids were identified from multiple databases and were collected using Venny 2.1. Microarray data of MCF-7 and MCF-7/MTX cells were acquired from the Gene Expression Omnibus. Subsequently, we constructed a protein-protein interaction network and selected the hub proteins. Gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, drug- and disease-gene enrichment analyses, genetic alteration examination, receiver operating characteristic curve analysis, mRNA levels analysis, prognostic value analysis, and molecular docking analysis were performed along with in vitro experiments. Cytotoxicity of citrus flavonoids (individually and combined) was assessed in MCF-7/MTX cells. Nobiletin and sinensetin significantly enhanced the cytotoxicity of MTX in MCF-7/MTX cells. BCL2L1, CDK1, EGFR, PTGS2, PLK1, MMP2, ACHE, ABCG2, and KIT genes were enriched in cholinesterase activity, cell cycle regulation, and the PI3K/Akt signaling pathway. Nobiletin and sinensetin impeded PLK1, CDK1, and ACHE activities based on molecular docking. Nobiletin and sinensetin in combination with MTX may overcome breast cancer cell resistance to MTX.
Collapse
Affiliation(s)
- Adam Hermawan
- Laboratory of Macromolecular Engineering, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia.
- Laboratory of Advanced Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Naufa Hanif
- Doctoral Student, School of Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
| | - Dyaningtyas Dewi Pamungkas Putri
- Laboratory of Advanced Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Laboratory of Pharmacology and Toxicology, Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Nurul Fatimah
- Laboratory of Advanced Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Heri Himawan Prasetio
- Laboratory of Macromolecular Engineering, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
661
|
Damrath M, Döring A, Nachtsheim BJ. Halogen bond-catalyzed Pictet-Spengler reaction. Chem Commun (Camb) 2025; 61:4828-4831. [PMID: 40034016 DOI: 10.1039/d4cc06635a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
We report an efficient halogen bond-catalyzed Pictet-Spengler reaction using diaryliodonium salts as catalysts as a metal-free alternative to traditional acid catalysis. Through systematic optimization, exceptional catalytic activity was achieved with only 0.5 mol% of a simple dibenzoiodolium with a perfluorinated borate counterion. The protocol demonstrates a broad substrate scope, converting various N-protected tryptamines and diverse carbonyl compounds (aromatic, heteroaromatic, and aliphatic aldehydes) into the corresponding tetrahydro-β-carbolines (THβCs) in up to 98% yield. The reaction versatility was further demonstrated by a successful oxa-variant using tryptophol. Control experiments revealed the crucial role of halogen bonding in ensuring efficient reaction progress.
Collapse
Affiliation(s)
- Mattis Damrath
- Institute for Organic and Analytical Chemistry, University of Bremen, 28359 Bremen, Germany.
| | - Alessandra Döring
- Institute for Organic and Analytical Chemistry, University of Bremen, 28359 Bremen, Germany.
| | - Boris J Nachtsheim
- Institute for Organic and Analytical Chemistry, University of Bremen, 28359 Bremen, Germany.
| |
Collapse
|
662
|
Krishnan S, Roy A, Wong L, Gromiha M. DRLiPS: a novel method for prediction of druggable RNA-small molecule binding pockets using machine learning. Nucleic Acids Res 2025; 53:gkaf239. [PMID: 40173014 PMCID: PMC11963762 DOI: 10.1093/nar/gkaf239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/16/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
Ribonucleic Acid (RNA) is the central conduit for information transfer in the cell. Identifying potential RNA targets in disease conditions is a challenging task, given the vast repertoire of functional non-coding RNAs in a human cell. A potential druggable target must satisfy several criteria, including disease association, cellular accessibility, binding pockets for drug-like molecules, and minimal cross-reactivity. While several methods exist for prediction of druggable proteins, they cannot be repurposed for RNAs due to fundamental differences in their binding modality. Taking all these constraints into account, a new structure-based model, Druggable RNA-Ligand binding Pocket Selector (DRLiPS), is developed here to predict binding site-level druggability of any given RNA target. A novel strategy for sampling negative binding sites in RNA structures using three parallel approaches is demonstrated here to improve model specificity: backbone motif search, exhaustive pocket prediction, and blind docking. An external blind test dataset has also been curated to showcase the model's generalizability to both experimental and modelled apo state RNA structures. DRLiPS has achieved an F1-score of 0.70, precision of 0.61, specificity of 0.89, and recall of 0.73 on this external test dataset, outperforming two existing methods, DrugPred_RNA and RNACavityMiner. Further analysis indicates that the features selected for model-building generalize well to both apo and holo states with a backbone RMSD tolerance of 3 Å. It can also predict the effect of binding site single point mutations on druggability, which can aid in optimizing synthetic RNA aptamers for small molecule recognition. The DRLiPS model is freely accessible at https://web.iitm.ac.in/bioinfo2/DRLiPS/.
Collapse
Affiliation(s)
- Sowmya Ramaswamy Krishnan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
- TCS Research (Life Sciences division), Tata Consultancy Services, Hyderabad 500081, India
| | - Arijit Roy
- TCS Research (Life Sciences division), Tata Consultancy Services, Hyderabad 500081, India
| | - Limsoon Wong
- Department of Computer Science, National University of Singapore, 117417, Singapore
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
- Department of Computer Science, National University of Singapore, 117417, Singapore
| |
Collapse
|
663
|
Mader LK, Keillor JW. Methods for kinetic evaluation of reversible covalent inhibitors from time-dependent IC 50 data. RSC Med Chem 2025:d5md00050e. [PMID: 40162199 PMCID: PMC11951164 DOI: 10.1039/d5md00050e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/17/2025] [Indexed: 04/02/2025] Open
Abstract
Potent reversible covalent inhibitors are often slow in establishing their covalent modification equilibrium, resulting in time-dependent inhibition. While these inhibitors are commonly assessed using IC50 values, there are no methods available to analyze their time-dependent IC50 data to provide their inhibition (K i and ) and covalent modification rate (k 5 and k 6) constants, leading to difficulty in accurately ranking drug candidates. Herein, we present an implicit equation that can estimate these constants from incubation time-dependent IC50 values and a numerical modelling method, EPIC-CoRe, that can fit these kinetic parameters from pre-incubation time-dependent IC50 data. The application of these new methods is demonstrated by the evaluation of a known inhibitor, saxagliptin, providing results consistent with those obtained by other known methods. This work introduces two new practical methods of evaluation for time-dependent reversible covalent inhibitors, allowing for rigorous characterization to enable the fine-tuning of their binding and reactivity.
Collapse
Affiliation(s)
- Lavleen K Mader
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa Ontario K1N 6N5 Canada
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa Ontario K1N 6N5 Canada
| |
Collapse
|
664
|
Rihtar E, Fink T, Lebar T, Lainšček D, Kolenc Ž, Polajnar LK, Jerala R. Ligand-induced assembly of antibody variable fragments for the chemical regulation of biological processes. Cell Chem Biol 2025; 32:474-485.e5. [PMID: 39952240 PMCID: PMC11935766 DOI: 10.1016/j.chembiol.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/30/2024] [Accepted: 01/26/2025] [Indexed: 02/17/2025]
Abstract
Precise control of biological processes by the application of small molecules can increase the safety and efficiency of therapies. Adverse side effects of small molecule signals and/or immunogenicity of regulatory domains hinder their biomedical utility. Here, we designed small molecule-responsive switches, based on the conditional reassembly of human antibody variable fragments, called Fv-CID switches. The principle was validated using high-affinity antibodies against nicotine and β-estradiol to construct chemically responsive transcription factors. Further, we developed an Fv-CID switch responsive to bio-inert, clinically approved compound fluorescein, which was used to control the activity of chimeric antigen receptor (CAR) T cells and bispecific T cell engagers (BiTEs) in vivo. This study provides a framework to regulate the expression of endogenous genes, combine multiple chemical signals, and regulate T cell-based immunotherapy in an animal cancer model using a clinically approved small molecule regulator that could be customized for regulating therapeutic proteins or cells.
Collapse
Affiliation(s)
- Erik Rihtar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Tina Fink
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Tina Lebar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Živa Kolenc
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Lucija Kadunc Polajnar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; Centre for Technologies of Gene and Cell Therapy, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| |
Collapse
|
665
|
Gao W, Han X, Li L, Xu Y, Xu M, Gao Z, Wang C. Functionalized ZIF-8 as a versatile platform for drug delivery and cancer therapy: strategies, challenges and prospects. J Mater Chem B 2025; 13:3758-3785. [PMID: 40019146 DOI: 10.1039/d4tb02289k] [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: 03/01/2025]
Abstract
This review discusses the functionalization strategies of ZIF-8 and challenges and future developments in ZIF-8-based platforms for drug delivery and cancer therapy. We systematically evaluate a series of innovative ZIF-8 functionalization methods, including atomic doping, introduction of targeting molecules, and biomimetic mineralization technology, to achieve precise drug release. These functionalization strategies significantly enhance the targeted delivery and controlled release properties of ZIF-8, broaden the diversity of drug delivery systems, maximize therapeutic effects, and minimize systemic toxicity. In addition, this review explores the important role of ZIF-8 in tumor therapy. Its ability to encapsulate multiple therapeutic agents and its responsiveness to the tumor microenvironment significantly improve the therapeutic effect and reduce the side effects of traditional treatments. By integrating multiple therapeutic agents and performing surface modification, ZIF-8-based platforms may provide personalized and efficient treatment options for drug-resistant or recurrent cancers. This review also comprehensively discusses the synthesis methods, drug loading capacity, and potential clinical applications of ZIF-8, emphasizing the need to optimize its large-scale production and reproducibility. In addition, further studies on the long-term biocompatibility and biodegradability of ZIF-8-based systems are essential to ensure their safety in long-term treatment. In summary, this review highlights the structural advantages and significant therapeutic potential of ZIF-8 and calls for the transition of ZIF-8 from laboratory research to clinical application to provide more targeted, efficient, and friendly cancer treatment options.
Collapse
Affiliation(s)
- Wenyue Gao
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Xinping Han
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Ling Li
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Yan Xu
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Min Xu
- Chengdu Third People's Hospital, Chengdu 610031, China
| | - Zhu Gao
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| | - Cuijuan Wang
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, P. R. China
| |
Collapse
|
666
|
Borisov VB, Forte E. Carbon Monoxide and Prokaryotic Energy Metabolism. Int J Mol Sci 2025; 26:2809. [PMID: 40141451 PMCID: PMC11942997 DOI: 10.3390/ijms26062809] [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: 02/21/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Carbon monoxide (CO) plays a multifaceted role in both physiology and pathophysiology. At high levels, it is lethal to humans due to its tight binding to globins and cytochrome c oxidase. At low doses, CO can exhibit beneficial effects; it serves as an endogenous signaling molecule and possesses antibacterial properties, which opens up possibilities for its use as an antimicrobial agent. For this purpose, research is in progress to develop metal-based CO-releasing molecules, metal-free organic CO prodrugs, and CO-generating hydrogel microspheres. The energy metabolism of prokaryotes is a key point that may be targeted by CO to kill invading pathogens. The cornerstone of prokaryotic energy metabolism is a series of membrane-bound enzyme complexes, which constitute a respiratory chain. Terminal oxidases, at the end of this chain, contain hemes and are therefore potential targets for CO. However, this research area is at its very early stage. The impact of CO on bacterial energy metabolism may also provide a basis for biotechnological applications in which this gas is present. This review discusses the molecular basis of the effects of CO on microbial growth and aerobic respiration supported by different terminal oxidases in light of recent findings.
Collapse
Affiliation(s)
- Vitaliy B. Borisov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory, 119991 Moscow, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskie Gory, 119991 Moscow, Russia
| | - Elena Forte
- Department of Biochemical Sciences, Sapienza University of Rome, I-00185 Rome, Italy;
| |
Collapse
|
667
|
Torres SW, Lan C, Harthorn A, Schmitz Z, Blanchard PL, Collins J, Hackel BJ. Molecular Determinants of Affinity and Isoform Selectivity in Protein─Small Molecule Hybrid Inhibitors of Carbonic Anhydrase. Bioconjug Chem 2025; 36:549-562. [PMID: 40030409 DOI: 10.1021/acs.bioconjchem.5c00006] [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: 03/20/2025]
Abstract
Multiple studies have demonstrated the benefit of engineering hybrid ligands that combine the unique benefits of small molecules and proteins or peptides. However, the molecular complexity of hybrid ligands generates a parameter space so large it cannot be exhaustively explored. We systematically evaluated the impact of one molecular design element, conjugation site, on the discovery of functional protein-small molecule hybrids (PriSMs). We utilized a library of yeast-displayed fibronectin domain variants with amino acid and loop length diversity in the paratope and a single cysteine at one of 18 possible conjugation sites. The protein variants were coupled with maleimide-functionalized acetazolamide and sorted via competitive flow cytometry to discover potent and selective inhibitors of three isoforms of carbonic anhydrase. Deep sequencing of the resultant populations of functional PriSMs revealed an isoform-dependent distribution of conjugation site preferences. The top PriSMs showed potency and selectivity gains up to 23- and 100-fold (in this case, for CA-II vs CA-XII, with a 43-fold selectivity gain for CA-II vs CA-IX) relative to PEG2-acetazolamide alone. The presented study expands our fundamental understanding of the role of conjugation site in PriSM function and informs future PriSM engineering efforts by highlighting the benefit of conjugation site diversity in PriSM libraries.
Collapse
Affiliation(s)
- Sarah W Torres
- Department of Biomedical Engineering, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Crystal Lan
- Department of Chemical Engineering and Materials Science, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Abbigael Harthorn
- Department of Biomedical Engineering, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Zachary Schmitz
- Department of Chemical Engineering and Materials Science, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Paul L Blanchard
- Department of Chemical Engineering and Materials Science, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Jon Collins
- Department of Biomedical Engineering, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Benjamin J Hackel
- Department of Biomedical Engineering, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
- Department of Chemical Engineering and Materials Science, University of Minnesota─Twin Cities, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
668
|
Zuo Q, Song X, Yan J, Bao G, Li Y, Shen J, He Z, Hu K, Sun W, Wang R. Triazination/IEDDA Cascade Modular Strategy Installing Pyridines/Pyrimidines onto Tyrosine Enables Peptide Screening and Optimization. J Am Chem Soc 2025; 147:9576-9589. [PMID: 39885681 DOI: 10.1021/jacs.4c17615] [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: 02/01/2025]
Abstract
Modular chemical postmodification of peptides is a promising strategy that supports the optimization and innovation of hit peptide therapeutics by enabling rapid derivatization. However, current methods are primarily limited to traditional bio-orthogonal strategies and chemical ligation techniques, which require the preintroduction of non-natural amino acids and impose fixed methods that limit peptide diversity. Here, we developed the Tyrosine-1,2,3-Triazine Ligation (YTL) strategy, which constructs novel linkages (pyridine and pyrimidine) through a "one-pot, two-step" process combining SNAr and IEDDA reactions, promoting modular post modification of Tyr-containing peptides. After optimizing the YTL strategy and establishing standard procedures, we successfully applied it to the solid-phase postmodification of various biorelated peptides, such as the synthesis of dual-mode imaging probes and long-acting GLP-1 analogs. As a proof of concept, a library of 384 amphipathic peptides was constructed using YTL based on 96-well microfiltration plates. Modular modifications were then performed on the screened template tripeptide RYR, leading to the generation of 20 derivatives. The antibacterial activity of these derivatives was systematically characterized, identifying Z8 as a potential antibacterial candidate.
Collapse
Affiliation(s)
- Quan Zuo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Xinyi Song
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Jie Yan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Guangjun Bao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Yiping Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Jieting Shen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Zeyuan He
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Wangsheng Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences and Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 199 West Donggang Road, Lanzhou, Gansu 730000, P. R. China
| |
Collapse
|
669
|
Al Nasr IS, Ma J, Khan TA, Koko WS, Ben Abdelmalek I, Schobert R, van de Sande W, Biersack B. Antiparasitic and Antifungal Activities of Cetyl-Maritima, a New N-Cetyl-Modified Maritima Derivative. Antibiotics (Basel) 2025; 14:321. [PMID: 40149131 PMCID: PMC11939259 DOI: 10.3390/antibiotics14030321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: New drugs are urgently needed for the treatment of neglected tropical diseases including leishmaniasis and eumycetoma, as well as globally occurring parasitic diseases such as toxoplasmosis. Fragrances, both natural and synthetic, were shown to be a rich source for the development of new anti-infectives and warrant deeper investigations. Exemplarily, we synthetically optimized the fragrance 4-(4,8-dimethyl-3,7-nonadienyl)-pyridine, a.k.a. Maritima, a pyridine derivative with marine odor. Methods: A new cationic N-cetyl-modified derivative of Maritima (dubbed Cetyl-Maritima), obtained by alkylation of Maritima, was tested for its activity against Madurella mycetomatis (M. mycetomatis) fungi, as well as against Toxoplasma gondii (T. gondii) and Leishmania major (L. major) protozoal parasites. Results: Cetyl-Maritima was found to be more strongly antifungal than the parent Maritima and a known antibiotic cetylpyridinium salt. Cetyl-Maritima also showed a similar activity against T. gondii parasites and, most notably, exhibited sub-micromolar activity against L. major amastigotes. Conclusions: The considerable antileishmanial activity of Cetyl-Maritima might lead to the development of a new potent and cost-effective drug candidate for the therapy of leishmaniasis and other infectious diseases caused by kinetoplastid parasites.
Collapse
Affiliation(s)
- Ibrahim S. Al Nasr
- Department of Biology, College of Science, Qassim University, Buraydah 51452, Saudi Arabia; (I.S.A.N.); (W.S.K.); (I.B.A.)
| | - Jingyi Ma
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (J.M.); (W.v.d.S.)
| | - Tariq A. Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Waleed S. Koko
- Department of Biology, College of Science, Qassim University, Buraydah 51452, Saudi Arabia; (I.S.A.N.); (W.S.K.); (I.B.A.)
| | - Imen Ben Abdelmalek
- Department of Biology, College of Science, Qassim University, Buraydah 51452, Saudi Arabia; (I.S.A.N.); (W.S.K.); (I.B.A.)
| | - Rainer Schobert
- Organic Chemistry Laboratory, University Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany;
| | - Wendy van de Sande
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (J.M.); (W.v.d.S.)
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University Bayreuth, Universitätsstrasse 30, 95440 Bayreuth, Germany;
| |
Collapse
|
670
|
Alrouji M, Alshammari MS, Majrashi TA, Zuberi A, Shahwan M, Atiya A, Shamsi A. Unraveling human transferrin-tryptamine interactions: a computational and biophysical approach to Alzheimer's disease therapeutics. Front Pharmacol 2025; 16:1540736. [PMID: 40176911 PMCID: PMC11962429 DOI: 10.3389/fphar.2025.1540736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Neurodegeneration is a progressive loss of neurons that leads to affected cognitive and motor functions and is characterized by neurodegenerative disorders (NDs). Human transferrin (Htf) is a blood plasma glycoprotein that binds to iron and regulates the free iron in biological fluids. Free iron is a potent neurotoxin associated with the generation of Reactive oxygen species (ROS) and is ultimately linked to oxidative stress and neuronal damage. Thus, targeting iron homeostasis is an attractive strategy for the management of NDs, viz. Alzheimer's disease (AD). Tryptamine (Trp) is a naturally occurring monoamine, that has demonstrated promising roles in AD therapeutics. The present study aims to delineate the binding mechanism of Trp with Htf employing computational and spectroscopic approaches. Molecular docking ascertained the vital residues governing the Htf-Trp complex formation. Further, Molecular dynamic (MD) studies ascertained the structural dynamics and stability of the complex, implying that the binding of Trp causes minimal structural alterations in Htf, suggestive of the stability of the complex. The results from fluorescence spectroscopy demonstrated the binding of Trp with Htf with a binding constant (K) of 0.48 × 106 M-1, validating the in silico observations. This study provides a platform to understand the binding mechanism that may lead to novel therapeutic approaches targeting AD.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Mohammed S. Alshammari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Taghreed A. Majrashi
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Azna Zuberi
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Moyad Shahwan
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Akhtar Atiya
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King Khalid University (KKU), Muhayil, Asir, Saudi Arabia
| | - Anas Shamsi
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
671
|
Laurent C, Poncet G, Herskovits T, Alves de Sousa R, Le Corre L, Al-Azzani M, Koenig A, Birman S, Outeiro TF, Mansuy D, Dairou J. Inhibition of the Parkinson's Disease-Related Protein DJ-1 by Endogenous Neurotoxins of the 1,2,3,4-Tetrahydroisoquinoline Family. ACS Chem Neurosci 2025; 16:1034-1042. [PMID: 40009035 DOI: 10.1021/acschemneuro.4c00559] [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] [Indexed: 02/27/2025] Open
Abstract
The protein DJ-1 appears to play a protective role in the development of Parkinson's disease (PD). Here, we show that endogenous neurotoxins of the 1,2,3,4-tetrahydroisoquinoline family (TIQs), formed upon reaction of various aldehydes such as methylglyoxal (MGO) with the neurotransmitter dopamine, act as irreversible inhibitors of the esterase activity of human DJ-1, with IC50 values between 15 and 57 μM. The presence of a catechol function appears to be essential for these inhibitory effects, which may be at the origin of the oxidation of cysteine 106, a crucial residue in the DJ-1 active site, thereby leading to DJ-1 inhibition. We also show that these endogenous neurotoxins inhibit the protective effects of DJ-1 against glycated guanosine diphosphate (GDP) formation and against alpha-synuclein (aSyn) aggregation induced by MGO. In total, the observed inhibition of DJ-1 by these endogenous neurotoxins may contribute to their damaging effects on the nervous system and, should be taken into account in therapeutic strategies for PD and related disorders.
Collapse
Affiliation(s)
- Catherine Laurent
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, 45 rue des Saints Pères, F-75006 Paris, France
| | - Gabrielle Poncet
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, 45 rue des Saints Pères, F-75006 Paris, France
- Genes Circuits Rhythms and Neuropathology team, Brain Plasticity unit, CNRS, ESPCI Paris - PSL, 75005 Paris, France
| | - Tristan Herskovits
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, 45 rue des Saints Pères, F-75006 Paris, France
| | - Rodolphe Alves de Sousa
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, 45 rue des Saints Pères, F-75006 Paris, France
| | - Laurent Le Corre
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, 45 rue des Saints Pères, F-75006 Paris, France
| | - Mohammed Al-Azzani
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Waldweg 33, 37073 Göttingen, Germany
| | - Annekatrin Koenig
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Waldweg 33, 37073 Göttingen, Germany
| | - Serge Birman
- Genes Circuits Rhythms and Neuropathology team, Brain Plasticity unit, CNRS, ESPCI Paris - PSL, 75005 Paris, France
| | - Tiago Fleming Outeiro
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Waldweg 33, 37073 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, U.K
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Straße 3a, 37075 Göttingen, Germany
| | - Daniel Mansuy
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, 45 rue des Saints Pères, F-75006 Paris, France
| | - Julien Dairou
- Université Paris Cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, 45 rue des Saints Pères, F-75006 Paris, France
| |
Collapse
|
672
|
Haque A, Alenezi KM, Abdul Rasheed MSM, Rahman MA, Anwar S, Ahamad S, Gupta D. Experimental and theoretical studies on structural changes in the microtubule affinity-regulating kinase 4 (MARK4) protein induced by N-hetarenes: a new class of therapeutic candidates for Alzheimer's disease. Front Med (Lausanne) 2025; 12:1529845. [PMID: 40177269 PMCID: PMC11962044 DOI: 10.3389/fmed.2025.1529845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/18/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Alzheimer's disease (AD) is a neurodegenerative disorder that progressively affects the cognitive function and memory of the affected person. Unfortunately, only a handful of effective prevention or treatment options are available today. Microtubule affinity-regulating kinase 4 (MARK4) is a serine/threonine protein that plays a critical role in regulating microtubule dynamics and facilitating cell division. The dysregulated expression of MARK4 has been associated with a range of diseases, including AD. Methods In this study, we synthesized a series of N-hetarenes via Pd(0)-catalyzed Suzuki-Miyaura cross coupling reaction. All compounds were characterized using multi-spectroscopic techniques and evaluated for their activity against the MARK4 enzyme through ATPase inhibition assays. The experimental data was further supported by computational and quantum chemical calculations. We also computed the drug-likeness, bioavailability, and toxicity (ADME/T) profiles of the compounds. Results Six new 4-(6-(arylpyrimidin-4-yl)piperazine-1-carboximidamides 5-10 were prepared in good yields. ATPase inhibition assay conducted on these compounds demonstrated IC50 values in micromolar range (5.35 ± 0.22 to 16.53 ± 1.71 μM). Among the tested compounds, 4-(6-(p-tolyl)pyrimidin-4-yl)piperazine-1-carboximidamide (5; IC50 = 5.35 ± 0.22 μM) and 4-(6-(benzo[b]thiophen-2-yl)pyrimidin-4-yl)piperazine-1-carboximidamide (9; IC50 = 6.68 ± 0.80 μM) showed the best activity. The binding constant (K), as determined by the fluorescence quenching assay was estimated to be 1.5 ± 0.51 × 105 M-1 for 5 and 1.14 ± 0.26 × 105 M-1 for 9. The results of molecular docking and MD simulation studies against MARK4 (PDB: 5ES1) indicated that compounds were able to bind the ATP binding pocket of the MARK4, leading to its stabilization. Additionally, ADME/T analysis revealed a high degree of drug-likeness of the compounds. Conclusion We demonstrated that 4-(6-(arylpyrimidin-4-yl)piperazine-1-carboximidamides) are a promising class of N-hetarenes for developing next-generation anti-AD drugs. The reported class of compounds inhibited MARK4 activity in-vitro at micromolar concentration by targeting the ATP-binding pocket. These findings provide valuable insights for future drug design.
Collapse
Affiliation(s)
- Ashanul Haque
- Department of Chemistry, College of Science, University of Hail, Hail, Saudi Arabia
| | - Khalaf M. Alenezi
- Department of Chemistry, College of Science, University of Hail, Hail, Saudi Arabia
| | | | - Md. Ataur Rahman
- Chemistry Program, New York University Abu Dhabi (NYUAD), Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Shahzaib Ahamad
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| |
Collapse
|
673
|
Oduselu GO, Ajani OO, Ogunnupebi TA, Elebiju OF, Bodun DS, Opebiyi OT, Adebiyi E. Synthesis, in silico and in vitro antimicrobial efficacy of some amidoxime-based benzimidazole and benzimidamide derivatives. RSC Med Chem 2025:d5md00114e. [PMID: 40162201 PMCID: PMC11950986 DOI: 10.1039/d5md00114e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025] Open
Abstract
Amidoximes are employed as building blocks to synthesise heterocyclic motifs with biological significance. They are very reactive and are used as prodrugs of amidine. This present study unveils the synthesis of amidoxime-based benzimidazole and benzimidamide motifs and evaluates their in silico and in vitro antimicrobial potential as future drug candidates. The compounds (2a, 2b, 4a-c) were synthesized using multi-step synthetic pathways. The synthesised compounds were characterised using physico-chemical examination, 1H- and 13C-NMR, DEPT-135, and FT-IR spectroscopic analyses. The in silico antimicrobial potentials of the synthesized compounds were carried out against glucosamine-6-phosphate synthase of E. coli (PDB ID: 2VF5), and N-myristoyltransferase (NMT) of C. albicans (PDB ID: 1IYL), while the in vitro antimicrobial screening was investigated against selected bacteria and fungi. The in silico studies were carried out using predicted ADMET screening, molecular docking, MM-GBSA, induced-fit docking (IFD), and molecular dynamics (MD) simulation studies. Furthermore, the in vitro experimental validations were performed using the agar diffusion method and the standard antibacterial and antifungal drugs used were gentamicin and ketoconazole respectively. The predicted toxicity test of the compounds showed no significant risk, except for 4c, which showed high tumorigenic risk. Compounds 2b and 2a gave better binding energies; -8.0 kcal mol-1 for 2VF5 and -11.7 kcal mol-1 for 1IYL, respectively. The antimicrobial zone of inhibition and minimum inhibitory concentration values were 40 mm and 3.90 mg mL-1 against S. mutans, then 42 mm and 1.90 mg mL-1 against C. albicans. Potential antimicrobial drug candidates have been identified in this report and should be explored for future preclinical research.
Collapse
Affiliation(s)
- Gbolahan O Oduselu
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana P. O Box LG 54, Legon Accra Ghana
| | - Olayinka O Ajani
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- Department of Chemistry, Covenant University Km 10 Idiroko Road P.M.B. 1023 Ota Ogun State Nigeria
| | - Temitope A Ogunnupebi
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- Department of Chemistry, Covenant University Km 10 Idiroko Road P.M.B. 1023 Ota Ogun State Nigeria
| | - Oluwadunni F Elebiju
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- Department of Chemistry, Covenant University Km 10 Idiroko Road P.M.B. 1023 Ota Ogun State Nigeria
| | - Damilola S Bodun
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
| | | | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University Ota Nigeria
- African Center of Excellence in Bioinformatics and Data Intensive Science, Makerere University Kampala Uganda
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ) Heidelberg Germany
| |
Collapse
|
674
|
Moosavi F, Divar M, Khabnadideh S, Tavakkoli M, Mohabbati M, Saso L, Poustforoosh A, Firuzi O. Spiroindoline quinazolinedione derivatives as inhibitors of P-glycoprotein: potential agents for overcoming multidrug resistance in cancer therapy. Mol Divers 2025:10.1007/s11030-025-11150-5. [PMID: 40106127 DOI: 10.1007/s11030-025-11150-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
Multidrug resistance (MDR) presents a major challenge for effectiveness of chemotherapy. This study investigates the effectiveness of spiroindoline quinazolinediones in reversing MDR mediated by P-glycoprotein (P-gp) overexpression in cancer cells. A series of synthesized hybrid spiro[indoline-3,2'-quinazoline]-2,4'(3'H)-dione derivatives (compounds 5a-5l) were analyzed for their ability to enhance rhodamine 123 (Rhd123) accumulation in the MES-SA/DX5 cell line using flow cytometry. The MTT assay was also employed to evaluate the compounds' effectiveness in reversing drug resistance. Additionally, docking studies and molecular dynamics simulations were conducted to investigate the interaction of these compounds with the P-gp transporter. The Rhd123 accumulation assay in MDR cancer cells revealed that most compounds, in particular 5f, 5g, 5h, 5i, 5j, 5k, and 5l, exhibited significant potential as P-gp inhibitors. Among the tested derivatives, compounds 5g and 5l demonstrated the best effects, and increased Rhd123 accumulation up to 12.9 times compared to untreated cells. Additionally, compounds 5f through 5 l bearing methylbenzyl (5f), benzyl (5g), pentyl (5 ), p-bromobenzyl (5i), p-chlorobenzyl (5j), dichlorobenzyl (5k), and tert-butylbenzyl (5l) substituents on the isatin ring effectively restored sensitivity to doxorubicin at their non-toxic concentrations in resistant MES-SA/DX5 cells. Among these, compound 5l at 5 μM exhibited the highest inhibitory potential, and lowered doxorubicin's IC50 value 10.1 times compared to control. Moreover, in silico investigation identified the potential interactions of test compounds with critical residues of P-gp involved in its efflux function. Our study suggests that the synthesized spiroindoline quinazolinediones may have high potentials as agents capable of reversing MDR in cancer cells.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoumeh Divar
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soghra Khabnadideh
- School of Pharmacy, Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marjan Tavakkoli
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Mohabbati
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. Le Aldo Moro 5, 00185, Rome, Italy
| | - Alireza Poustforoosh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
675
|
Das S, Babu NK, Mazire P, Roy A, Kumar R, Singh S, Sharma DK. Indolylmaleimide derivatives as a new class of anti-leishmanial agents: synthesis and biological evaluation. RSC Med Chem 2025:d5md00132c. [PMID: 40256310 PMCID: PMC12004262 DOI: 10.1039/d5md00132c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/09/2025] [Indexed: 04/22/2025] Open
Abstract
Leishmaniasis is a neglected tropical disease, primarily affecting poor and developing countries. The present therapeutic approach faces various limitations, such as concerns regarding toxicity, route of administration, and the emergence of drug resistance. Therefore, there is a critical need to identify novel scaffolds to combat this fatal parasitic infection. Leishmanial DNA topoisomerase 1B is a heterodimeric protein and plays a crucial role in resolving topological problems during various biological processes. It is structurally distinct from its human counterparts, making it an attractive target for drug discovery. In this study, we synthesized various aminated indolylmaleimide derivatives targeting the leishmanial topoisomerase 1B enzyme. In vitro leishmanicidal assays on Leishmania promastigotes identified one highly potent hit (3m), showing considerable inhibition with single-digit micromolar IC50 values. Moreover, molecular docking analysis of the potent hit (3m) confirmed its strong binding affinity with the enzyme. Thus, the hit molecule (3m) holds promise as a lead for developing novel therapeutic strategies against leishmaniasis.
Collapse
Affiliation(s)
- Samarpita Das
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University Varanasi-221005 India
| | - Neerupudi Kishore Babu
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) Sector-67, S.A.S. Nagar (Mohali)-160062 India
| | - Priyanka Mazire
- Department of Biotechnology, Savitribai Phule Pune University Pune-411007 India
| | - Amit Roy
- Department of Biotechnology, Savitribai Phule Pune University Pune-411007 India
| | - Rohit Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University Varanasi-221005 India
| | - Sushma Singh
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) Sector-67, S.A.S. Nagar (Mohali)-160062 India
| | - Deepak K Sharma
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University Varanasi-221005 India
| |
Collapse
|
676
|
Zhu Q, Hu L, Cui C, Zang M, Dong H, Ma J. Decoding Hairpin Structure Stability in Lin28-Mediated Repression. Biochemistry 2025; 64:1276-1284. [PMID: 40020242 DOI: 10.1021/acs.biochem.4c00675] [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: 03/19/2025]
Abstract
The Lin28 protein is well known for its role in inhibiting the biogenesis of microRNAs (miRNAs) that belong to the let-7 family. The Lin28 and let-7 axes are associated with several types of cancers. It is imperative to understand the underlying mechanism to treat these cancers in a more efficient way. In this study, we employed all-atom molecular dynamics simulation as a research tool to investigate the interaction formed between Lin28 and the precursor element of let-7d, one of the 12 members of the let-7 family. By constructing systems of an intact sequence length of preE-let-7d, our simulations suggest that both the loop region of the hairpin structure and the GGAG sequence can form stable interactions with the cold shock domain (CSD) and zinc knuckle domain (ZKD) regions of the protein, respectively. The system, by deleting the nucleotides GGAG at the 3' terminal, indicates that the loop region is more responsible for its ability in bypassing the binding and repression of Lin28. Additionally, using let-7c-2, which can bypass Lin28 regulation, as a template, we constructed systems with mutated loop region sequences in miRNAs and tested their stabilities. Our simulation results coincide well with experimental observations. Based on both simulation results and statistical analysis from two databases, we hypothesized that two factors, namely, the interaction between terminal nucleotides and the ring tension originating from the middle nucleotides, can significantly influence their stabilities. Systems combining strong and weak terminal interactions with large and small ring tensions were recruited to validate our hypothesis. Our findings offer a new perspective and shed light on strategies for designing sequences to regulate the interactions formed between proteins and hairpin structures.
Collapse
Affiliation(s)
- Qiang Zhu
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Limu Hu
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Chang Cui
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Min Zang
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, P. R. China
| | - Hao Dong
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center (ChemBIC), & Institute for Brain Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jing Ma
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
677
|
Goonetilleke EC, Huang X. Targeting Bacterial RNA Polymerase: Harnessing Simulations and Machine Learning to Design Inhibitors for Drug-Resistant Pathogens. Biochemistry 2025; 64:1169-1179. [PMID: 40014017 PMCID: PMC12016775 DOI: 10.1021/acs.biochem.4c00751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The increase in antimicrobial resistance presents a major challenge in treating bacterial infections, underscoring the need for innovative drug discovery approaches and novel inhibitors. Bacterial RNA polymerase (RNAP) has emerged as a crucial target for antibiotic development due to its essential role in transcription. RNAP is a molecular motor, and its function relies heavily on the dynamic shifts between multiple conformational states. While biochemical and structural experimental methods offer crucial insights into static RNAP-drug interactions, they fall short in capturing the dynamics at a molecular level. By integrating experimental data with advanced computational techniques like Markov State Models (MSMs), Generalized Master Equation (GME) Models and other machine-learning models constructed from molecular dynamics (MD) simulations, researchers can elucidate novel cryptic pockets that open transiently for antibiotic compounds and gain a more nuanced and comprehensive understanding of RNAP-drug interactions. This integrated approach not only deepens our fundamental knowledge but also enables more targeted and efficient antibiotic design strategies. In this Perspective, we highlight how this synergy between experimental and computational methods has the potential to open new pathways for innovative drug design and combination therapies that may help turn the tide in the ongoing battle against antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Eshani C. Goonetilleke
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
| | - Xuhui Huang
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
678
|
Wang M, Huang X, Zhang D, Liu Y, Liu P. The role of fructose-1,6-bisphosphatase 1 on regulating the cancer progression and drug resistance. Discov Oncol 2025; 16:346. [PMID: 40100307 PMCID: PMC11920503 DOI: 10.1007/s12672-025-02112-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
Fructose-1,6-bisphosphatase 1 (FBP1) is the enzyme that limits the process of gluconeogenesis as it facilitates the hydrolysis of fructose-1,6-bisphosphate(F-1,6-BP) to produce fructose-6-phosphate(F6P) and inorganic phosphate. Gluconeogenesis is the production of glucose from small carbohydrate substrates. The gluconeogenic process is typically suppressed in cancer because it inhibits glycolysis. Apart from its involvement in cellular glucose metabolism, FBP1 also plays a role in gene transcription, mRNA translation and stability regulation, and the immune microenvironment of tumors. Because of its multifaceted functions, the mechanisms by which FBP1 is involved in tumor development are complex. Moreover, FBP1 deficiency is associated with radiation and chemotherapy resistance and poor prognosis in cancer patients. Restoration of FBP1 expression in cancer cells is expected to hold promise for cancer therapy. However, up to now few reviews have systematically summarized the important functional mechanisms of FBP1 in tumorigenesis and the small molecule compounds that restore FBP1 expression. Therefore, this article addresses the question "How does FBP1 contribute to cancer progression, and can targeting FBP1 be a potential therapeutic approach?" by summarizing the effects of FBP1 on cancer development and progression as well as its mediated drug resistance and the future clinical applications of potential small molecule modulators targeting FBP1.
Collapse
Affiliation(s)
- Mengmeng Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Xiaoju Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Dan Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Yisan Liu
- Department of Urology, People's Hospital of Cili, Cili, 427200, Hunan, China.
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China.
| |
Collapse
|
679
|
Zerfas BL, Liu Y, Che J, Donovan KA, Hatcher JM, Huerta F, Metivier RJ, Nowak RP, Ragosta L, Tsang T, Fischer ES, Jones LH. Structure-guided design of a truncated heterobivalent chemical probe degrader of IRE1α. RSC Med Chem 2025:d5md00028a. [PMID: 40151563 PMCID: PMC11938282 DOI: 10.1039/d5md00028a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025] Open
Abstract
IRE1α is an ER protein involved in the unfolded protein response (UPR) and dysregulation of the ER stress pathway has been implicated in several diseases. Inhibitors of the cytoplasmic endonuclease or kinase domains of the enzyme have limited utility and targeted degradation would address additional scaffolding functions of the protein. Here, we describe the design and development of IRE1α proteolysis targeting chimeras (PROTACs) based on a lysine-reactive salicylaldehyde RNase inhibitor, and present the structure-activity relationships (SARs) that delivered the first highly selective degraders of a native ER-membrane associated protein. Medicinal chemistry optimization exploited ternary complex computational modelling to inform design, HiBiT-SpyTag IRE1α degradation and NanoBRET cereblon occupancy cell-based assays to generate SARs, and mass spectrometry-based proteomics to assess broad selectivity in an unbiased manner. Merging IRE1α and CRBN ligand chemotypes provided the truncated chimera CPD-2828 with physicochemical properties more akin to an oral molecular glue degrader than a traditional PROTAC.
Collapse
Affiliation(s)
- Breanna L Zerfas
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA USA
| | - Yingpeng Liu
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA USA
| | - Jianwei Che
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA USA
| | - Katherine A Donovan
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA USA
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA USA
| | - John M Hatcher
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA USA
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA USA
| | - Fidel Huerta
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA USA
| | - Rebecca J Metivier
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA USA
| | - Radosław P Nowak
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA USA
| | - Leah Ragosta
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA USA
| | - Tiffany Tsang
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA USA
| | - Eric S Fischer
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA USA
- Department of Cancer Biology, Dana-Farber Cancer Institute Boston MA USA
| | - Lyn H Jones
- Center for Protein Degradation, Dana-Farber Cancer Institute Boston MA USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School Boston MA USA
| |
Collapse
|
680
|
Le Stum M, Romero E, Molander GA. Photocatalyzed elaboration of antibody-based bioconjugates. Beilstein J Org Chem 2025; 21:616-629. [PMID: 40130177 PMCID: PMC11931643 DOI: 10.3762/bjoc.21.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
Antibody-drug conjugates (ADCs) represent a promising class of targeted therapeutics, combining the specificity of antibodies with the potency of cytotoxic drugs to enhance therapeutic efficacy while minimizing off-target effects. The development of new chemical methods for bioconjugation is essential to generate ADCs and to optimize their stability, efficacy, and safety. Traditional conjugation methods often face challenges related to site-selectivity and heterogeneous product mixtures, highlighting the need to develop new, innovative chemical strategies. Photoredox chemistry emerges as a powerful tool in this context, enabling precise, mild, and selective modifications of peptides and proteins. By harnessing light to drive chemical transformations, photoredox techniques can facilitate the synthesis of antibody bioconjugates. This perspective will discuss the drive to develop and empower photoredox methods applied to antibody functionalization.
Collapse
Affiliation(s)
- Marine Le Stum
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-Sur-Yvette, France
| | - Eugénie Romero
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-Sur-Yvette, France
| | - Gary A Molander
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-Sur-Yvette, France
| |
Collapse
|
681
|
Marton J, Cumming P, Rice KC, Linders JTM. Morphinan Alkaloids and Their Transformations: A Historical Perspective of a Century of Opioid Research in Hungary. Int J Mol Sci 2025; 26:2736. [PMID: 40141378 PMCID: PMC11943231 DOI: 10.3390/ijms26062736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
The word opium derives from the ancient Greek word ὄπιον (ópion) for the juice of any plant, but today means the air-dried seed capsule latex of Papaver somniferum. Alkaloid chemistry began with the isolation of morphine from crude opium by Friedrich Wilhelm Adam Sertürner in 1804. More than a century later, Hungarian pharmacist János Kabay opened new perspectives for the direct isolation of morphine from dry poppy heads and straw without the labor-intensive harvesting of opium. In 2015, Kabay's life and achievements obtained official recognition as constituting a «Hungarikum», thereby entering the national repository of matters of unique cultural value. To this day, the study of Papaver alkaloids is a focus of medicinal chemistry, the (perhaps unstated) aspiration of which is to obtain an opioid with lesser abuse potential and side effects, while retaining good analgesic properties. We begin this review with a brief account of opiate biosynthesis, followed by a detailed presentation of semisynthetic opioids, emphasizing the efforts of the Alkaloida Chemical Company, founded in 1927 by János Kabay, and the morphine alkaloid group of the University of Debrecen.
Collapse
Affiliation(s)
- János Marton
- ABX Advanced Biochemical Compounds Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Strasse 10-14, D-01454 Radeberg, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Freiburgstraße 18, CH-3010 Bern, Switzerland;
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Kenner C. Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, NIDA and the NIAAA, NIH, Department of Health and Human Services, 9800 Medical Center Drive, Bethesda, MD 20892, USA;
| | | |
Collapse
|
682
|
López-Fernández AM, Neto JC, de Llanos R, Miravet JF, Galindo F. Minimalistic bis-triarylpyridinium cations: effective antimicrobials against bacterial and fungal pathogens. RSC Med Chem 2025:d4md00902a. [PMID: 40190417 PMCID: PMC11969996 DOI: 10.1039/d4md00902a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/17/2025] [Indexed: 04/09/2025] Open
Abstract
A series of twelve compounds from the family of 2,4,6-triarylpyridinium cations have been synthesized, chemically characterized (1H, 13C NMR, HRMS), and microbiologically evaluated (MIC determination against S. aureus, E. faecalis, E. coli, P. aeruginosa, and C. albicans). These compounds are quaternary ammonium cations (QACs), classified as either mono-QACs or bis-QACs. The mono-QACs are further divided into those with short (three-carbon) and long (twelve-carbon) pendant chains. An additional structural variable is the number of bromine atoms attached to the aromatic rings, ranging from zero to three. The major findings of this study are: (a) bis-QACs exhibit notably higher antimicrobial activity than mono-QACs; (b) an increased number of bromine atoms on the structure appears to diminish antimicrobial properties and (c) one of the compounds (1a) shows particularly promising properties as a broad spectrum antimicrobial, given its low MICs across all five pathogenic microorganisms studied. Preliminary assays with C. albicans show that 1a has a strong mitochondrial activity, causing a remarkable mitochondrial membrane depolarization in this organelle. Taken together, this study positions triarylpyridinium cations-previously unexplored as antimicrobials-as promising candidates for future drug development, especially in light of the growing concern over drug-resistant microorganisms.
Collapse
Affiliation(s)
- Ana M López-Fernández
- Departamento de Química Inorgánica y Orgánica, Universitat Jaume I Av. V. Sos Baynat s/n 12071 Castellón Spain
| | - Jean C Neto
- Departamento de Química Inorgánica y Orgánica, Universitat Jaume I Av. V. Sos Baynat s/n 12071 Castellón Spain
| | - Rosa de Llanos
- Unidad Predepartamental de Medicina, Universitat Jaume I Av. V. Sos Baynat s/n 12071 Castellón Spain
| | - Juan F Miravet
- Departamento de Química Inorgánica y Orgánica, Universitat Jaume I Av. V. Sos Baynat s/n 12071 Castellón Spain
| | - Francisco Galindo
- Departamento de Química Inorgánica y Orgánica, Universitat Jaume I Av. V. Sos Baynat s/n 12071 Castellón Spain
| |
Collapse
|
683
|
Liu Z, Yang Y, Xie X, Li R, You J, Zhao X, Wang Y, Guo J. Development and Characterization of LasR Immobilized Monolithic Column for Screening Active Ingredients as Quorum Sensing Inhibitors Against P. aeruginosa in Natural Products. Drug Des Devel Ther 2025; 19:2051-2064. [PMID: 40124560 PMCID: PMC11929421 DOI: 10.2147/dddt.s501621] [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: 10/18/2024] [Accepted: 03/01/2025] [Indexed: 03/25/2025] Open
Abstract
Background and Aim The enzyme/protein immobilized monolithic capillary combined with liquid chromatography-mass spectrometry is an efficient screening strategy for the corresponding agonist/antagonist. LasR is the potential therapeutic target since it plays a vital role in the colonization and invasion of Pseudomonas aeruginosa (P. aeruginosa). Therefore, reagents that inhibit LasR may be effective against P. aeruginosa. To screen and find LasR inhibitors rapidly, a LasR-immobilized monolithic capillary column was prepared and characterized. Methods Firstly, the recombinant LasR protein was prepared in E. coli. Then, the LasR protein was immobilized to the surface of poly (glycidyl methacrylate-co-poly(ethylene glycol)diacrylate)-ethylenediamine monolithic column. The affinity and stability of prepared column was also evaluated. Furthermore, the prepared column was applied to fishing LasR inhibitor in Scutellaria baicalensis Georgi extract. The interaction of the screening compound to LasR was confirmed through molecular docking. Results The recombinant active LasR protein was prepared in E. coli. After purification and validation, a comparative ligand fishing monolithic column was prepared through immobilizing LasR to the surface of the poly (glycidyl methacrylate-co-poly(ethylene glycol)diacrylate)-ethylenediamine through amidation reaction. The LasR was successfully immobilized to the monolithic column characterizing by Fourier transform infrared spectroscopy and scanning electron microscopy. The activity of immobilized LasR was reserved as it has affinity to the nature ligand 3-oxo-C12-HSL and stablied within 24 h at 4 °C. In the Scutellaria baicalensis Georgi extract, baicalein was screened as a potential LasR inhibitor. The molecular docking results and the in vivo evaluation confirmed the activity of baicalein. Conclusion The proposed LasR immobilized monolithic column is a viable strategy in screening LasR inhibitors. It can be considered as a possible alternative to traditional methods for screening LasR inhibitors as drug candidates against P. aeruginosa.
Collapse
Affiliation(s)
- Zheng Liu
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Yue Yang
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Xiaoyuan Xie
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Rui Li
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Jifeng You
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Xianglong Zhao
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
- College of Pharmacy, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Yuanyuan Wang
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
| | - Jialiang Guo
- School of Medicine, Foshan University, Foshan, 528000, People’s Republic of China
- College of Pharmacy, Jinan University, Guangzhou, 510632, People’s Republic of China
- Guangdong Provincial Engineering Technology Research Center of Whole Process Quality Control and Analysis of Lingnan Traditional Chinese Medicine, Foshan, 528225, People’s Republic of China
| |
Collapse
|
684
|
Adebayo G, Ayanda OI, Rottmann M, Ajibaye OS, Oduselu G, Mulindwa J, Ajani OO, Aina O, Mäser P, Adebiyi E. The Importance of Murine Models in Determining In Vivo Pharmacokinetics, Safety, and Efficacy in Antimalarial Drug Discovery. Pharmaceuticals (Basel) 2025; 18:424. [PMID: 40143200 PMCID: PMC11944934 DOI: 10.3390/ph18030424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
New chemical entities are constantly being investigated towards antimalarial drug discovery, and they require animal models for toxicity and efficacy testing. Murine models show physiological similarities to humans and are therefore indispensable in the search for novel antimalarial drugs. They provide a preclinical basis (following in vitro assessments of newly identified lead compounds) for further assessment in the drug development pipeline. Specific mouse strains, non-humanized and humanized, have successfully been infected with rodent Plasmodium species and the human Plasmodium species, respectively. Infected mice provide a platform for the assessment of treatment options being sought. In vivo pharmacokinetic evaluations are necessary when determining the fate of potential antimalarials in addition to the efficacy assessment of these chemical entities. This review describes the role of murine models in the drug development pipeline. It also explains some in vivo pharmacokinetic, safety, and efficacy parameters necessary for making appropriate choices of lead compounds in antimalarial drug discovery. Despite the advantages of murine models in antimalarial drug discovery, certain limitations are also highlighted.
Collapse
Affiliation(s)
- Glory Adebayo
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Department of Biological Sciences, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
- Biochemistry and Nutrition Division, Nigerian Institute of Medical Research, Yaba PMB 2013, Nigeria; (O.S.A.); (O.A.)
| | - Opeyemi I. Ayanda
- Department of Biological Sciences, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
| | - Matthias Rottmann
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland; (M.R.); (P.M.)
| | - Olusola S. Ajibaye
- Biochemistry and Nutrition Division, Nigerian Institute of Medical Research, Yaba PMB 2013, Nigeria; (O.S.A.); (O.A.)
| | - Gbolahan Oduselu
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Department of Chemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
| | - Julius Mulindwa
- Department of Biochemistry and Sports Science, College of Natural Sciences, Makerere University, Kampala P.O. Box 7062, Uganda;
| | - Olayinka O. Ajani
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Department of Chemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Nigeria
| | - Oluwagbemiga Aina
- Biochemistry and Nutrition Division, Nigerian Institute of Medical Research, Yaba PMB 2013, Nigeria; (O.S.A.); (O.A.)
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, CH-4123 Allschwil, Switzerland; (M.R.); (P.M.)
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota PMB 1023, Nigeria; (G.A.); (G.O.); (O.O.A.)
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- African Centre of Excellence in Bioinformatics & Data Intensive Science (ACE), Kampala P.O. Box 7062, Uganda
- Infectious Diseases Institute, Makerere University, Kampala P.O. Box 22418, Uganda
| |
Collapse
|
685
|
Taheri-Khas Z, Gharzi A, Vaissi S, Heshmatzad P, Kalhori Z. Advanced sperm preservation techniques in yellow spotted mountain newts Neurergus derjugini enhance genetic management and conservation efforts. Sci Rep 2025; 15:9334. [PMID: 40102525 PMCID: PMC11920051 DOI: 10.1038/s41598-025-93284-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
Advances in cold storage and cryopreservation of amphibian sperm are critical for the genetic management and conservation of threatened species. This study represents the first investigation into the sperm of the yellow spotted mountain newt (Neurergus derjugini), focusing on both short-term and long-term storage for future reproductive efforts. We examined the effects of seven extenders on sperm motility over time at three storage temperatures (4 ± 1 °C, 9 ± 1 °C, and 20 ± 1 °C). Additionally, we assessed the impact of 16 cryoprotectants on sperm motility and morphology post-thawing. Following the identification of the most effective freezing medium, we evaluated sperm DNA fragmentation to ensure viability. Our results indicate that 10% Holtfreter's solution is the optimal extender for short-term storage at all three temperatures, maintaining sperm motility for up to 15 days at 4 °C. For long-term storage, a combination of 10% Holtfreter's solution and 10% DMSO was found to best preserve sperm motility, morphology, and minimize DNA fragmentation after thawing. These findings underscore the importance of specific extenders and temperature treatments in enhancing sperm functionality, thereby supporting successful assisted reproductive technologies (ART) for endangered species.
Collapse
Affiliation(s)
- Zeynab Taheri-Khas
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Ahmad Gharzi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran.
| | - Somaye Vaissi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran.
| | - Pouria Heshmatzad
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Zahra Kalhori
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| |
Collapse
|
686
|
Moustafa SMN, Elkanzi NAA, Bakr RB. Synthesis and Evaluating the Anticandidal Activities of Triazino[4,3-a]Quinolinecarboxylate Derivatives: A Promising Approach to Combat Candida Infections. Chem Biodivers 2025:e202500045. [PMID: 40100043 DOI: 10.1002/cbdv.202500045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/20/2025]
Abstract
This study aimed to synthesize novel triazino[4,3-a]quinolinecarboxylate compounds (4, 6, 8, and 10) and evaluate these compounds for their antifungal activity against Candida species. Compound 8 was a standout candidate which demonstrated superior efficacy against C. albicans (minimum inhibitory concentration [MIC] = 45 µg/mL), C. glabrata (MIC = 32 µg/mL), C. parapsilosis (MIC = 36 µg/mL) and C. guilliermondii (MIC = 32 µg/mL) compared to Miconazole (MIC = 50-60 µg/mL). Furthermore, the induced morphological and ultra-structural changes by compound 8 on Candida cells are analyzed by light and transmission electron microscopy. Significant alterations in the viability and the architecture of Candida cells highlight the potential of compound 8 as a lead for further use as an antifungal drug. SARs displayed that substitution with the cyano group (as in compounds 8 and 10) was critical for anticandida potency. A molecular docking study of the most active compounds 8 and 10 was conducted inside 14α-demethylase (CYP51) to predict the binding mode of these compounds as antifungal. The most active compound 8 (MIC = 32-45 µg/mL) demonstrated the highest binding energy score which is equal to -8.93 kcal/mol. The findings of the in vitro anticandidal potential have been supported by molecular docking studies.
Collapse
Affiliation(s)
- Shaima M N Moustafa
- Biology Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Nadia A A Elkanzi
- Chemistry Department, College of Science, Jouf University, Sakaka, Saudi Arabia
| | - Rania B Bakr
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
687
|
Kagho MD, Schmidt K, Lambert C, Jia L, Venkatakrishnan V, Mehr L, Bylund J, Rottner K, Stadler M, Stradal TEB, Klahn P. NQO1-Responsive Prodrug for in Cellulo Release of Cytochalasin B as Cancer Cell-Targeted Migrastatic. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2410861. [PMID: 40095397 DOI: 10.1002/smll.202410861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Migrastatic drugs targeting cell motility and suppressing invasiveness of solid tumors, have the potential to bring about a paradigm shift in the treatment of solid cancer. Cytochalasin B (CB) is a potent migrastatic compound, but its clinical use is limited by poor selectivity. Here, a NQO1-responsive prodrug, BQTML-CB is developed, synthesized in three steps from cytochalasin B derived from Preussia similis G22. BQTML-CB is selectively activated in NQO1-positive cancer cells, releasing active CB. In vitro, BQTML-CB significantly inhibits proliferation and migration in NQO1-positive U-2OS cells, causing actin disruption and cytokinesis abnormalities, while sparing NQO1-negative B16-F1 cells. The prodrug shows reduced effects on human neutrophils, indicating reduced immunosuppressive activity of BQTML-CB compared to CB. Co-culture studies reveal a beneficial bystander effect, as cleaved CB diffused into adjacent NQO1-deficient cells. These findings support BQTML-CB as a cancer-targeted prodrug with selective antiproliferative and migrastatic properties, highlighting the potential of C7-OH-modified cytochalasans in cancer therapy.
Collapse
Affiliation(s)
- Mervic D Kagho
- Department of Chemistry and Molecular Biology, Division of Organic and Medicinal Chemistry, University of Gothenburg, Natrium, Medicinaregatan 7B, Gothenburg, 413 90, Sweden
| | - Katharina Schmidt
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Christopher Lambert
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany
- Department of Microbial Drugs, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Lili Jia
- Department of Microbial Drugs, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Vignesh Venkatakrishnan
- Department of Life Sciences, Chalmers University of Technology, Kemivägen 4, Göteborg, 41296, Sweden
| | - Luisa Mehr
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Johan Bylund
- Department of Oral Microbiology and Immunology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 12F, Göteborg, 413 90, Sweden
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany
| | - Marc Stadler
- Department of Microbial Drugs, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
- Institute for Microbiology, Technische Universität Braunschweig, Spielmannstraße 7, 38106, Braunschweig, Germany
| | - Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Philipp Klahn
- Department of Chemistry and Molecular Biology, Division of Organic and Medicinal Chemistry, University of Gothenburg, Natrium, Medicinaregatan 7B, Gothenburg, 413 90, Sweden
| |
Collapse
|
688
|
Chong CSC, Lau YY, Michels PAM, Lim CSY. Insights into biofilm-mediated mechanisms driving last-resort antibiotic resistance in clinical ESKAPE pathogens. Crit Rev Microbiol 2025:1-26. [PMID: 40098357 DOI: 10.1080/1040841x.2025.2473332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025]
Abstract
The rise of antibiotic-resistant bacteria poses a grave threat to global health, with the ESKAPE pathogens, which comprise Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. being among the most notorious. The World Health Organization has reserved a group of last-resort antibiotics for treating multidrug-resistant bacterial infections, including those caused by ESKAPE pathogens. This situation calls for a comprehensive understanding of the resistance mechanisms as it threatens public health and hinder progress toward the Sustainable Development Goal (SDG) 3: Good Health and Well-being. The present article reviews resistance mechanisms, focusing on emerging resistance mutations in multidrug-resistant ESKAPE pathogens, particularly against last-resort antibiotics, and describes the role of biofilm formation in multidrug-resistant ESKAPE pathogens. It discusses the latest therapeutic advances, including the use of antimicrobial peptides and CRISPR-Cas systems, and the modulation of quorum sensing and iron homeostasis, which offer promising strategies for countering resistance. The integration of CRISPR-based tools and biofilm-targeted approaches provides a potential framework for managing ESKAPE infections. By highlighting the spread of current resistance mutations and biofilm-targeted approaches, the review aims to contribute significantly to advancing our understanding and strategies in combatting this pressing global health challenge.
Collapse
Affiliation(s)
- Christina Shook Cheng Chong
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, No 1, Jalan UCSI, UCSI Heights, Taman Connaught, Cheras, Kuala Lumpur, Malaysia
| | - Yin Yin Lau
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, No 1, Jalan UCSI, UCSI Heights, Taman Connaught, Cheras, Kuala Lumpur, Malaysia
| | - Paul A M Michels
- School of Biological Sciences, University of Edinburgh, The King's Buildings, Edinburgh 3FL, UK
| | - Crystale Siew Ying Lim
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, No 1, Jalan UCSI, UCSI Heights, Taman Connaught, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
689
|
Jaen Maisonet I, Sharafi M, Korchak EJ, Salazar-Chaparro A, Bratt A, Parikh T, Varca AC, Shah B, Darnowski M, Chung M, Teh WP, Che J, Bezsonova I, Buhrlage SJ. Small-molecule allosteric activator of ubiquitin-specific protease 7 (USP7). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643379. [PMID: 40161813 PMCID: PMC11952563 DOI: 10.1101/2025.03.14.643379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Ubiquitin-specific protease 7 (USP7) is a deubiquitylase essential for cell homeostasis, DNA repair, and regulation of both tumor suppressors and oncogenes. Inactivating USP7 mutations have been associated with Hao-Fountain Syndrome (HAFOUS), a rare neurodevelopmental disorder. Although a range of USP7 inhibitors have been developed over the last decade, in the context of HAFOUS as well as oncogene regulation, USP7 activators may represent a more relevant approach. To address this challenge, we report the discovery and characterization of a small-molecule activator of USP7 called MS-8. We showed that MS-8 activates USP7 by engaging the allosteric C-terminal binding pocket of USP7, thus mimicking the allosteric autoactivation by the USP7 C-terminal tail. We observed that MS-8 engages and activates mutant USP7 in a cellular context, impacting downstream proteins. Taken together, our study provides validation of the USP7 activator that paves the way towards novel activation-driven USP7 pharmacology.
Collapse
|
690
|
Song ZJ, Wu XF, Zhou ZY, Zhang JJ, Pan YY, Dong X, Pang X, Xie YP, Sun J, Zhang Y, Qin J. Design, synthesis, and evaluation of carboxylic acid-substituted celecoxib isosteres as potential anti-inflammatory agents. Eur J Med Chem 2025; 286:117286. [PMID: 39854941 DOI: 10.1016/j.ejmech.2025.117286] [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: 12/03/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
A library comprising twenty-four isosteric derivatives of celecoxib substituted with carboxylic acid (labeled as 5a-5x), was synthesized and characterized through 1H NMR, 13C NMR, HRMS, and elemental analysis. Molecular docking studies revealed that all compounds successfully docked into the binding pocket of COX-2, and the introduction of carboxyl group enhances the interaction between the derivatives and COX-2. The compounds were further evaluated for cell toxicity, and in vitro anti-inflammatory activity. Notably, compound 5l exhibited significant inhibition of both COX-2 and NO release in vitro in comparison to the standard compound, displaying the highest selectivity towards the COX-2 enzyme (SI = 295.9) in comparison to celecoxib (SI = 261.3). 5l also exhibited the most potent anti-inflammatory activity and safety (ulcer index = 5.2) in vivo comparable to celecoxib at the same concentration. Through the molecular modeling and dynamics analysis, it was observed that compound 5l effectively stabilized within the active binding site of COX-2 through strong hydrogen bond interactions, and through the ADMET studies investigated the physiochemical properties and drug-likeliness behavior of compound 5l. In conclusion, compound 5l demonstrated to be a potential selective COX-2 anti-inflammatory candidate with reduced gastrointestinal risks.
Collapse
Affiliation(s)
- Zi-Jie Song
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, PR China
| | - Xiao-Fei Wu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, PR China
| | - Zhi-Ya Zhou
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, PR China
| | - Jing-Jing Zhang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, PR China
| | - Yan-Yan Pan
- Qilu Medical University, Zibo, 255300, PR China
| | - Xue Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, PR China
| | - Xuan Pang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, PR China
| | - Ya-Ping Xie
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, PR China
| | - Juan Sun
- School of Biological & Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, 310023, PR China.
| | - Yu Zhang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, PR China.
| | - Jie Qin
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, PR China.
| |
Collapse
|
691
|
Nia MH, Garzia L, Jawhar W, Wilson LD, van de Ven TGM. Bioinspired complex cellulose nanorod-architectures: A model for dual-responsive smart carriers. Carbohydr Polym 2025; 352:123162. [PMID: 39843067 DOI: 10.1016/j.carbpol.2024.123162] [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: 09/23/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 01/24/2025]
Abstract
The synergy between nanomaterials as solid supports and supramolecular concepts has resulted in nanomaterials with hierarchical structure and enhanced functionality. Herein, we developed and investigated innovative supramolecular functionalities arising from the synergy between organic moieties and the preexisting nanoscale soft material backbones. Based on these complex molecular nano-architectures, a new nanorod carbohydrate polymer carrier was designed with bifunctional hairy nanocellulose (BHNC) to reveal dual-responsive advanced drug delivery (ADD). This carrier scavenges K+-ions within cancer cells, while simultaneously releasing doxorubicin, combining ion homeostasis disruption with targeted drug delivery. The BC ADD system resulted from cross-linking dibenzo-18-crown-6-ether (DB18C6) with BHNC particles. To enhance cellular internalization and facilitate tracking of uptake, the cellulose nanorod carrier was labeled with biotin and fluorescein isothiocyanate, referred to as BCFB. The BHNC serves as the backbone, while the immobilized DB18C6 moieties can capture doxorubicin via complex formation. The BCFB complex molecular nanorod carriers exhibit distinct ADD profiles with pH and K+-responsiveness. They were evaluated as biocompatible carriers for ADD in MDA-MB-231 breast cancer cells, including quantification of nanoparticle uptake and flow cytometry with KHOS cells. These cellulose-based carriers possess unique structure and properties with potential utility as phase transfer catalysts and as adsorbents for diverse waterborne contaminants.
Collapse
Affiliation(s)
- Marzieh Heidari Nia
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, SK S7N 5C9, Canada; Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada; Quebec Centre for Advanced Materials (QCAM) and Pulp and Paper Research Centre, McGill University, 3420 University Street, Montreal, QC H3A 2A7, Canada.
| | - Livia Garzia
- Department of Surgery, Faculty of Medicine and Health Sciences, McGill University, Canada; Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| | - Wajih Jawhar
- Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| | - Lee D Wilson
- Department of Chemistry, University of Saskatchewan, 110 Science Place, Saskatoon, SK S7N 5C9, Canada.
| | - Theo G M van de Ven
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada; Quebec Centre for Advanced Materials (QCAM) and Pulp and Paper Research Centre, McGill University, 3420 University Street, Montreal, QC H3A 2A7, Canada.
| |
Collapse
|
692
|
Faucher FF, Lovell S, Bertolini M, Blažková K, Cosco ED, Bogyo M, Barniol-Xicota M. Macrocyclic phage display for identification of selective protease substrates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643185. [PMID: 40161698 PMCID: PMC11952493 DOI: 10.1101/2025.03.13.643185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Traditional methods for identifying selective protease substrates have primarily relied on synthetic libraries of linear peptides, which offer limited sequence and structural diversity. Here, we present an approach that leverages phage display technology to screen large libraries of chemically modified cyclic peptides, enabling the identification of highly selective substrates for a protease of interest. Our method uses a reactive chemical linker to cyclize peptides on the phage surface, while simultaneously incorporating an affinity tag and a fluorescent reporter. The affinity tag enables capture of the phage library and subsequent release of phages expressing optimal substrates upon incubation with a protease of interest. The addition of a turn-on fluorescent reporter allows direct quantification of cleavage efficiency throughout each selection round. The resulting identified substrates can then be chemically synthesized, optimized and validated using recombinant enzymes and cells. We demonstrate the utility of this approach using Fibroblast Activation Protein alpha (FAPα) and the related proline-specific protease, dipeptidyl peptidase-4 (DPP4), as targets. Phage selection and subsequent optimization identified substrates with selectivity for each target that have the potential to serve as valuable tools for applications in basic biology and fluorescence image-guided surgery (FIGS). Overall, our strategy provides a rapid and unbiased platform for effectively discovering highly selective, non-natural protease substrates, overcoming key limitations of existing methods.
Collapse
Affiliation(s)
- Franco F. Faucher
- Department of Chemistry, Stanford University, Stanford, CA 94305, United States
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Scott Lovell
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Life Sciences, University of Bath, Bath, BA2 7AX, United Kingdom
| | - Matilde Bertolini
- Department of Genetics, School of Medicine, Stanford University, California 94305, United States
| | - Kristýna Blažková
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Emily D. Cosco
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, United States
| | - Marta Barniol-Xicota
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, United States
- Department of Medicine and Life Sciences, Biomedical Research Park (PRBB), Universitat Pompeu Fabra, 08003 Barcelona
| |
Collapse
|
693
|
Zlatković Đ, Dobričić V, Srbljanović J, Lijeskić O, Bauman N, Ćirković V, Štajner T. N-(9-Acridinyl) Amino Acid Derivatives: Synthesis and In Vitro Evaluation of Anti- Toxoplasma gondii Activity. Pharmaceutics 2025; 17:374. [PMID: 40143037 PMCID: PMC11944910 DOI: 10.3390/pharmaceutics17030374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Acridine, an aromatic heterocyclic compound, serves as a basis for the synthesis of potent bioactive derivatives, displaying a broad spectrum of biological activity, such as antibacterial, antitumor, and antiparasitic activity. With the ability to undergo various types of electrophilic substitutions, introducing different side chains could lead to compounds being active towards various and potentially multiple biotargets. Toxoplasma gondii, a ubiquitous protozoan parasite with worldwide distribution, poses a major health threat, particularly in immunocompromised patients and fetuses. Current treatment options for toxoplasmosis are scarce, with notable limitations, especially regarding side myelotoxicity and inactivity towards T. gondii cysts, causing a need for novel drug candidates. The aim of this study was to evaluate selected N-(9-acrydinil) amino acid derivatives as potential anti-T. gondii agents. Methods: Synthesis of new derivatives was performed using a two-step method, with the initial mixing of 9-chloroacridine with methanol and sodium alkoxide solution and subsequent adding of appropriate amino acids. Cytotoxicity of the tested compounds was evaluated on the Vero cell line using a MTT assay, while their anti-T. gondii activity was investigated using T. gondii RH strain tachyzoites. Results: CC50 values of the derivatives ranged from 41.72 to 154.10 µM. Anti-T. gondii activity, displayed as a reduction in the number of viable tachyzoites compared to the untreated control, ranged from 0 to 33.3%. One of the derivatives displayed activity comparable to the standard treatment option while retaining acceptable cytotoxicity. Esterification, presence of aromatic substituents and the length of the amino acid side chain were identified as key factors that affect both toxicity and activity of these derivatives. Conclusions: Promising results obtained throughout this study provide guidelines for further structural modifications of N-(9-acrydinil) amino acid derivatives in order to synthesize drug candidates competitive to standard treatment options for toxoplasmosis.
Collapse
Affiliation(s)
- Đorđe Zlatković
- National Reference Laboratory for Toxoplasmosis, Group for Microbiology and Parasitology, Center of Excellence for Food- and Vector-born Zoonosis, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (Đ.Z.); (J.S.); (O.L.); (N.B.); (V.Ć.)
| | - Vladimir Dobričić
- Department of Pharmaceutical Chemistry, University of Belgrade-Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia;
| | - Jelena Srbljanović
- National Reference Laboratory for Toxoplasmosis, Group for Microbiology and Parasitology, Center of Excellence for Food- and Vector-born Zoonosis, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (Đ.Z.); (J.S.); (O.L.); (N.B.); (V.Ć.)
| | - Olivera Lijeskić
- National Reference Laboratory for Toxoplasmosis, Group for Microbiology and Parasitology, Center of Excellence for Food- and Vector-born Zoonosis, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (Đ.Z.); (J.S.); (O.L.); (N.B.); (V.Ć.)
| | - Neda Bauman
- National Reference Laboratory for Toxoplasmosis, Group for Microbiology and Parasitology, Center of Excellence for Food- and Vector-born Zoonosis, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (Đ.Z.); (J.S.); (O.L.); (N.B.); (V.Ć.)
| | - Vladimir Ćirković
- National Reference Laboratory for Toxoplasmosis, Group for Microbiology and Parasitology, Center of Excellence for Food- and Vector-born Zoonosis, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (Đ.Z.); (J.S.); (O.L.); (N.B.); (V.Ć.)
| | - Tijana Štajner
- National Reference Laboratory for Toxoplasmosis, Group for Microbiology and Parasitology, Center of Excellence for Food- and Vector-born Zoonosis, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (Đ.Z.); (J.S.); (O.L.); (N.B.); (V.Ć.)
| |
Collapse
|
694
|
Wagenaar GTM, Moll GN. Advances in the therapeutic potentials of ligands of the apelin receptor APJ. Eur J Pharmacol 2025; 991:177302. [PMID: 39870231 DOI: 10.1016/j.ejphar.2025.177302] [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: 10/02/2024] [Revised: 01/08/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Angiotensin II protein J receptor, APJ, is a type A G protein coupled receptor. Endogenous apelin and elabela peptides stimulate APJ via distinct signalling profiles. A complex signalling map of elabela-stimulated APJ was published in 2022. Dimerization or oligomerization of APJ with itself or other receptor(s) can affect APJ signalling. Apelin has been shown to tolerate mutations and/or modifications at multiple sites without abolishing activity. This offers a great opportunity to design and engineer variants with desired signalling profiles and enhanced resistance to breakdown by peptidases. Several biased agonists with enhanced therapeutic potential have been generated. APJ agonists have therapeutic potential in multiple diseases including cardiovascular, renal, pulmonary and metabolic diseases, and viral infections. APJ antagonists may have therapeutic potential in cancer and retinopathy, and in related diseases in which unwanted angiogenesis is to be halted. A growing understanding of APJ signalling pathways and the robust therapeutic potential of associated ligands for many serious diseases will stimulate the clinical development of APJ ligands.
Collapse
Affiliation(s)
- Gerry T M Wagenaar
- Division of VitalTissue, Multi Tissue Center ETB-BISLIFE, Jan van Krimpenweg 17, 2031 CG, Haarlem, the Netherlands
| | - Gert N Moll
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG, Groningen, the Netherlands.
| |
Collapse
|
695
|
An Y, Lv X, Xu S, Li H, Zheng P, Zhu W, Wang L. Pyrimidine-based dual-target inhibitors targeting epidermal growth factor receptor for overcoming drug resistance in cancer therapy(2006-present). Eur J Med Chem 2025; 286:117268. [PMID: 39837171 DOI: 10.1016/j.ejmech.2025.117268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
The epidermal growth factor receptor (EGFR) is a pivotal member of the epidermal growth factor receptor family, exerting crucial regulatory influence on cellular physiological processes, particularly in relation to cell growth, proliferation, and differentiation. In recent years, numerous EGFR inhibitors have been introduced to the market; unfortunately, the effectiveness of single-target EGFR inhibitors has been compromised due to the development of drug resistance caused by EGFR mutations. Despite attempts by some researchers to address this issue through combination therapy with two or more drugs, instances of dose-limiting toxicities have been observed. Consequently, EGFR dual-target inhibitors have emerged as a burgeoning field in cancer treatment, offering a novel therapeutic option for solid tumors with the added benefits of reduced risk of resistance, lower dosage requirements, diminished toxicity profiles, and enhanced efficacy. At present, a series of EGFR dual-target inhibitors with diverse structures have been developed successively. In this study, we initially investigated the pyrimidine-based EGFR dual-target inhibitors that have been reported in the past two decades and categorized them into aminopyrimidine derivatives and heterocyclic pyrimidine derivatives with increased molecular complexity. Subsequently, we comprehensively summarized the biological activity and structure-activity relationship of this class of inhibitors in the context of cancer therapy, while also exploring potential opportunities and challenges associated with their application in this field. The present study provides a partial framework to guide future endeavors in drug development.
Collapse
Affiliation(s)
- Yufeng An
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Xinya Lv
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Shidi Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Heqing Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| | - Linxiao Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi, 330013, China.
| |
Collapse
|
696
|
Jiang Y, Wang Y, Su F, Hou Y, Liao W, Li B, Mao W. Insights into NEK2 inhibitors as antitumor agents: From mechanisms to potential therapeutics. Eur J Med Chem 2025; 286:117287. [PMID: 39832390 DOI: 10.1016/j.ejmech.2025.117287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
NEK2, a serine/threonine protein kinase, is integral to mitotic events such as centrosome duplication and separation, microtubule stabilization, spindle assembly checkpoint, and kinetochore attachment. However, NEK2 overexpression leads to centrosome amplification and chromosomal instability, which are significantly associated with various malignancies, including liver, breast, and non-small cell lung cancer. This overexpression could facilitate tumor development and confer resistance to therapy by promoting aberrant cell division and centrosome amplification. Consequently, inhibiting NEK2 is considered as a promising strategy for oncological therapy. To date, no small molecule NEK2-specific inhibitors have advanced into clinical trials, highlighting the necessity for optimized design and the deployment of innovative technologies. In this review, we will provide a comprehensive summary of the chemical structure, biological functions, and disease associations of NEK2, focusing on the existing NEK2 small molecule inhibitors, especially their structure-activity relationships, limitations, and research strategies. Our objective is to provide valuable insights for the future development of NEK2 inhibitors and analysis of challenges faced in translating these findings into clinical applications.
Collapse
Affiliation(s)
- Yizhen Jiang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Healthand, Department of Frontiers Science Center for Disease-related Molecular Network, Core Facilities, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yutong Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Healthand, Department of Frontiers Science Center for Disease-related Molecular Network, Core Facilities, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Feijing Su
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Healthand, Department of Frontiers Science Center for Disease-related Molecular Network, Core Facilities, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yaqin Hou
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Wen Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Baichuan Li
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Healthand, Department of Frontiers Science Center for Disease-related Molecular Network, Core Facilities, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Wuyu Mao
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Healthand, Department of Frontiers Science Center for Disease-related Molecular Network, Core Facilities, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
697
|
Liu J, Zhang L, Ma H, Sun H, Ge SA, Liu J, Fan S, Quan C. Quaternary ammonium chitosan-functionalized mesoporous silica nanoparticles: A promising targeted drug delivery system for the treatment of intracellular MRSA infection. Carbohydr Polym 2025; 352:123184. [PMID: 39843087 DOI: 10.1016/j.carbpol.2024.123184] [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: 08/13/2024] [Revised: 11/28/2024] [Accepted: 12/22/2024] [Indexed: 01/30/2025]
Abstract
The limited membrane permeability and bacterial resistance pose significant challenges in the management of intracellular drug-resistant bacterial infections. To overcome this issue, we developed a bacterial-targeted drug delivery system based on quaternary ammonium chitosan-modified mesoporous silica nanoparticles (MSN-NH2-CFP@HACC) for the treatment of intracellular Methicillin-resistant Staphylococcus aureus (MRSA) infections. This system utilizes amino-functionalized mesoporous silica nanoparticles to efficiently load cefoperazone (CFP), and the nanoparticles' surface is coated with 2-hydroxypropyltrimethyl ammonium chloride chitosan (HACC) to target bacteria and enhance macrophage uptake. The findings indicate that MSN-NH2-CFP@HACC nanoparticles are efficiently internalized by macrophages, demonstrate accelerated drug release in acidic environments, and exhibit enhanced antibacterial properties, effectively suppressing the proliferation and intracellular escape of MRSA. Moreover, HACC enhances the bacterial capture ability of the nanoparticles and reduces resistance by disrupting bacterial membrane structures and inhibiting bacterial β-lactamase activity. In a murine model of MRSA bacteremia, MSN-NH2-CFP@HACC exhibited remarkable antibacterial efficacy and significantly attenuated severe inflammatory responses. In conclusion, MSN-NH2-CFP@HACC represent a promising antibiotic delivery system with exceptional antibacterial efficacy and favorable biocompatibility, thus presenting a novel strategy for addressing intracellular drug-resistant bacterial infections and demonstrating significant potential for clinical application.
Collapse
Affiliation(s)
- Junfeng Liu
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Liying Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Haodi Ma
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Haoyang Sun
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Shu-Ai Ge
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Jieyi Liu
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Shengdi Fan
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China
| | - Chunshan Quan
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian 116600, PR China; Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian 116600, PR China.
| |
Collapse
|
698
|
Pei G, Wang P, Lin L, Zhang H, Wei R, Liao S. Photocatalytic Radical Azido/Fluorosulfonylation of Unactivated Alkenes: Accessing Hubs Bridging CuAAC and SuFEx Click Chemistry. Org Lett 2025; 27:2467-2474. [PMID: 40017314 DOI: 10.1021/acs.orglett.5c00426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Herein, we describe the successful development of an azido-fluorosulfonylation reaction of alkenes under photoredox catalysis, which could allow the installation of the two "clickable" groups, -N3 and -SO2F, on a C-C double bond, with TMSN3 as the azide source. The utilization of the difunctionalization products is also demonstrated in the construction of a library of 1,2,3-triazolesulfonyl fluoride compounds as well as drug molecule ligation by merging copper-catalyzed azide-alkyne cycloaddition (CuAAC) and sulfur(VI) fluoride exchange (SuFEx), the two generations of click reactions. Mechanistic studies suggest a radical fluorosulfonylation/azidation mechanism and unveil FSO2N3 as a new and potential fluorosulfonyl radical precursor.
Collapse
Affiliation(s)
- Guanhua Pei
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Peng Wang
- Key Laboratory of Green and Precise Synthetic Chemistry and Application, Ministry of Education, College of Chemistry and Materials Science, Huaibei, Normal University, Huaibei, Anhui 235000, China
| | - Lu Lin
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Honghai Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Rongbiao Wei
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Saihu Liao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
699
|
Jin X, Zhang H, Xie X, Zhang M, Wang R, Liu H, Wang X, Wang J, Li D, Li Y, Xue W, Li J, He J, Liu Y, Yao J. From Traditional Efficacy to Drug Design: A Review of Astragali Radix. Pharmaceuticals (Basel) 2025; 18:413. [PMID: 40143189 PMCID: PMC11945149 DOI: 10.3390/ph18030413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Astragali Radix (AR), a traditional Chinese herbal medicine, is derived from the dried roots of Astragalus membranaceus (Fisch.) Bge. var. mongholicus (Bge.) Hsiao (A. membranaceus var. mongholicus, AMM) or Astragalus membranaceus (Fisch.) Bge (A. membranaceus, AM). According to traditional Chinese medicine (TCM) theory, AR is believed to tonify qi, elevate yang, consolidate the body's surface to reduce sweating, promote diuresis and reduce swelling, generate body fluids, and nourish the blood. It has been widely used to treat general weakness and chronic illnesses and to improve overall vitality. Extensive research has identified various medicinal properties of AR, including anti-tumor, antioxidant, cardiovascular-protective, immunomodulatory, anti-inflammatory, anti-diabetic, and neuroprotective effects. With advancements in technology, methods such as computer-aided drug design (CADD) and artificial intelligence (AI) are increasingly being applied to the development of TCM. This review summarizes the progress of research on AR over the past decades, providing a comprehensive overview of its traditional efficacy, botanical characteristics, drug design and distribution, chemical constituents, and phytochemistry. This review aims to enhance researchers' understanding of AR and its pharmaceutical potential, thereby facilitating further development and utilization.
Collapse
Affiliation(s)
- Xiaojie Jin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (X.J.); (H.Z.); (X.X.); (M.Z.); (X.W.); (J.W.)
- Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (Y.L.); (J.H.)
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China;
| | - Huijuan Zhang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (X.J.); (H.Z.); (X.X.); (M.Z.); (X.W.); (J.W.)
| | - Xiaorong Xie
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (X.J.); (H.Z.); (X.X.); (M.Z.); (X.W.); (J.W.)
| | - Min Zhang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (X.J.); (H.Z.); (X.X.); (M.Z.); (X.W.); (J.W.)
| | - Ruifeng Wang
- Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (Y.L.); (J.H.)
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China;
| | - Hao Liu
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (X.J.); (H.Z.); (X.X.); (M.Z.); (X.W.); (J.W.)
| | - Xinyu Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (X.J.); (H.Z.); (X.X.); (M.Z.); (X.W.); (J.W.)
| | - Jiao Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (X.J.); (H.Z.); (X.X.); (M.Z.); (X.W.); (J.W.)
| | - Dangui Li
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (X.J.); (H.Z.); (X.X.); (M.Z.); (X.W.); (J.W.)
| | - Yaling Li
- Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (Y.L.); (J.H.)
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Weiwei Xue
- Innovative Drug Research Centre, School of Pharmaceutical Sciences, Chongqing University, Chongqing 404100, China;
| | - Jintian Li
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China;
| | - Jianxin He
- Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (Y.L.); (J.H.)
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Yongqi Liu
- Provincial Key Laboratory of Molecular Medicine and Prevention Research of Major Diseases, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (Y.L.); (J.H.)
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China;
| | - Juan Yao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (X.J.); (H.Z.); (X.X.); (M.Z.); (X.W.); (J.W.)
- Key Laboratory of Dunhuang Medicine, Ministry of Education, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China;
| |
Collapse
|
700
|
Zhang Z, Li RP, Gong X, Xu X, Peng X, Tang S. Regioselective Pyrazole Synthesis via Base-Mediated [3+2] Cycloaddition of 2-Alkynyl-1,3-Dithianes and Sydnones. J Org Chem 2025; 90:3769-3778. [PMID: 40014796 DOI: 10.1021/acs.joc.5c00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
We present a novel base-mediated [3+2] cycloaddition for the regioselective synthesis of polysubstituted pyrazoles using 2-alkynyl-1,3-dithianes and sydnones. By exploiting the umpolung and nucleophilic properties of 2-alkynyl-1,3-dithianes, this method achieves efficient pyrazole construction under mild conditions with excellent regioselectivity, broad functional group tolerance, and diverse substrate compatibility. Furthermore, the unique reactivity of the dithianyl group enables facile derivatization and synthesis of highly functionalized pyrazoles.
Collapse
Affiliation(s)
- Zhuzhu Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P.R. China
| | - Rui-Peng Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P.R. China
| | - Xiaomeng Gong
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P.R. China
| | - Xiangrong Xu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P.R. China
| | - Xuan Peng
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P.R. China
| | - Shouchu Tang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P.R. China
- State Key Laboratory of Applied Organic Chemistry, Lanzhou University, Lanzhou 730000, P.R. China
| |
Collapse
|