751
|
U S, Basu U, Paira P. Current Context of Designing Phototheranostic Cyclometalated Iridium (III) Complexes to Open a New Avenue in Cancer Therapy. ChemMedChem 2025; 20:e202400649. [PMID: 39495110 DOI: 10.1002/cmdc.202400649] [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: 08/19/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Photo-induced chemotherapy offers the best option for the selective treatment of cancer among all the prevailing modalities. Iridium (III) complexes, flourished with excellent photophysical and photochemical properties, have been considered to be superior for undergoing photo-responsive cancer therapy. Large Stokes shift, long-lived triplet excited state, photostability, and tuneable emission have rendered its excellence as a phototheranostic agent. In particular, the cyclometalated Ir (III) complexes and their respective nanoparticles have made a strong niche in the arena of cancer therapy. In recent years, Ir (III) based complexes have shown promising utilities as both imaging and therapeutic agents as well. Therefore, this review summarises the recent advances in the strategic designing of cyclometalated Ir(III) complexes to augment their phototheranostic applications in precision medicine.
Collapse
Affiliation(s)
- Sreelekha U
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| | - Uttara Basu
- Department of Chemistry, Birla Institute of Technology & Science, Pilani K K Birla Goa Campus, NH 17 B, Zuarinagar, Goa, India
| | - Priyankar Paira
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| |
Collapse
|
752
|
Li H, Liu Y, Yuan H, Cai P, Wu T, Yang Z, Nie J, Zhang W, Huang Z, Liu N, Chen Y, Zhou Z. Development and Evaluation of Novel 68Ga/ 177Lu-Labeled PSMA Inhibitors with Enhanced Pharmacokinetics and Tumor Imaging for Prostate Cancer. Mol Pharm 2025; 22:1584-1597. [PMID: 39951557 DOI: 10.1021/acs.molpharmaceut.4c01302] [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: 02/16/2025]
Abstract
Prostate-specific membrane antigen (PSMA) has been a key target for diagnosing and treating prostate cancer, particularly in high-grade, metastatic, and therapy-resistant tumors. This study presents a series of novel 68Ga- and 177Lu-labeled PSMA inhibitors, derived from the previously developed [68Ga]Ga-Flu-1. We explored the impact of PEG chains, lipophilic macrocycles, and dimerization on their in vivo properties. The 68Ga- and 177Lu-labeled inhibitors were assessed for biodistribution and tumor targeting in PC3-PIP tumor xenografts, leading to the identification of several promising candidates based on imaging and tumor-specific uptake. Positron emission tomography (PET) imaging revealed that the poly(ethylene glycol)-modified [68Ga]Ga-BisPSMA-P4 demonstrated rapid tumor penetration and excellent tumor-to-background contrast. In comparative biodistribution studies, the naphthalene ring-modified [68Ga]Ga-BisPSMA-Nph-P4 showed higher tumor uptake (∼60% ID/g at 1 h postinjection) and rapid renal clearance (∼25% ID/g at 2 h postinjection). Additionally, [177Lu]Lu-BisPSMA-Nph-P4 displayed superior retention, with significant uptake on day 7, highlighting its potential as a novel PSMA inhibitor for prostate cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Haiyang Li
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Yang Liu
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Hongmei Yuan
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Ping Cai
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Tongtong Wu
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Zhicong Yang
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Jiaqi Nie
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Wei Zhang
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Zhanwen Huang
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Nan Liu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Yue Chen
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| | - Zhijun Zhou
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan 646000, China
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan 646000, China
| |
Collapse
|
753
|
Süssmuth RD, Kulike‐Koczula M, Gao P, Kosol S. Fighting Antimicrobial Resistance: Innovative Drugs in Antibacterial Research. Angew Chem Int Ed Engl 2025; 64:e202414325. [PMID: 39611429 PMCID: PMC11878372 DOI: 10.1002/anie.202414325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 11/30/2024]
Abstract
In the fight against bacterial infections, particularly those caused by multi-resistant pathogens known as "superbugs", the need for new antibacterials is undoubted in scientific communities and is by now also widely perceived by the general population. However, the antibacterial research landscape has changed considerably over the past years. With few exceptions, the majority of big pharma companies has left the field and thus, the decline in R&D on antibacterials severely impacts the drug pipeline. In recent years, antibacterial research has increasingly relied on smaller companies or academic research institutions, which mostly have only limited financial resources, to carry a drug discovery and development process from the beginning and through to the beginning of clinical phases. This review formulates the requirements for an antibacterial in regard of targeted pathogens, resistance mechanisms and drug discovery. Strategies are shown for the discovery of new antibacterial structures originating from natural sources, by chemical synthesis and more recently from artificial intelligence approaches. This is complemented by principles for the computer-aided design of antibacterials and the refinement of a lead structure. The second part of the article comprises a compilation of antibacterial molecules classified according to bacterial target structures, e.g. cell wall synthesis, protein synthesis, as well as more recently emerging target classes, e.g. fatty acid synthesis, proteases and membrane proteins. Aspects of the origin, the antibacterial spectrum, resistance and the current development status of the presented drug molecules are highlighted.
Collapse
Affiliation(s)
- Roderich D. Süssmuth
- Institut für ChemieTechnische Universität BerlinStrasse des 17. Juni 124, TC210629BerlinGermany
| | - Marcel Kulike‐Koczula
- Institut für ChemieTechnische Universität BerlinStrasse des 17. Juni 124, TC210629BerlinGermany
| | - Peng Gao
- Institut für ChemieTechnische Universität BerlinStrasse des 17. Juni 124, TC210629BerlinGermany
| | - Simone Kosol
- Medical School BerlinDepartment Human MedicineRüdesheimer Strasse 5014195BerlinGermany
| |
Collapse
|
754
|
Amin H, Kaur G, Goswami A, Bhat KA. Facile Synthesis of Ureidic Derivatives of Betulinic Acid With Antiproliferative Effects on Colorectal and Prostate Cancer Cells. Chem Biodivers 2025:e202402669. [PMID: 40025637 DOI: 10.1002/cbdv.202402669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/04/2025]
Abstract
Betulinic acid isolated from Platanus orientalis bark was modified in one-pot reaction to furnish 12 ureidic derivatives. The synthesized derivatives were tested against an array of cancer cell lines, colon HCT-116, breast MCF-7, pancreatic MIA PaCa-2, and prostate PC-3 cancer cell lines. Among the synthesized compounds, BA1 exhibited potent anti-cancer activity with IC50 of 0.84 and 3.87 µM against HCT-116 and PC-3 using doxorubicin as standard. Our study demonstrates the anti-proliferative and anti-metastatic role of BA1, thus inducing apoptosis in prostrate and colorectal cell lines.
Collapse
Affiliation(s)
- Henna Amin
- Bioorganic Chemistry Division, Indian Institute of Integrative Medicine (CSIR), Srinagar, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Gursimar Kaur
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Anindya Goswami
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Khursheed Ahmad Bhat
- Bioorganic Chemistry Division, Indian Institute of Integrative Medicine (CSIR), Srinagar, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
755
|
Nandakumar V, Selvi Ramasamy S, Adhigaman K, Arumugam D, Ramasamy S, Vivek R, Athimoolam S, Thangaraj S. Investigating the Antiproliferative Activity of Novel 4-Chloro-8-Nitro-1,2-Dihydro-3-Quinoline Acylhydrazones on Human Cervical Cancer Cell Lines. Chem Biodivers 2025; 22:e202401636. [PMID: 39543828 DOI: 10.1002/cbdv.202401636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/17/2024]
Abstract
A new series of acyl hydrazones have been synthesized from 4-chloro-8-nitro-1,2-dihydroquinoline-3-carbaldehyde. These compounds were characterized using various spectroscopic techniques. Density functional theoretical (DFT) studies were conducted to understand the correlation between electronic parameters and biological activity. The biological activity of the compounds was theoretically examined through molecular docking and ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) analysis. The compounds demonstrated high absorption rates and were found to be non-hepatotoxic. Preliminary cytotoxicity screenings against HeLa cell lines identified compound 7 as the most potent, with an IC50 value of 18.8 μM. This compound was further selected for bioimaging studies. The results indicate that compound 7 induces apoptosis at its IC50 concentration, suggesting its potential as an anticancer agent.
Collapse
Affiliation(s)
- Vandana Nandakumar
- Department of Chemistry, Bharathiar University, 641046, Coimbatore, Tamilnadu, India
| | | | - Kaviyarasu Adhigaman
- Department of Chemistry, Bharathiar University, 641046, Coimbatore, Tamilnadu, India
| | - Deepak Arumugam
- Department of Physics, Bharathiar University, 641046, Coimbatore, Tamilnadu, India
| | - Shankar Ramasamy
- Department of Physics, Bharathiar University, 641046, Coimbatore, Tamilnadu, India
| | - Raju Vivek
- Department of Zoology, Bharathiar University, 641046, Coimbatore, Tamilnadu, India
| | | | - Suresh Thangaraj
- Department of Chemistry, Bharathiar University, 641046, Coimbatore, Tamilnadu, India
| |
Collapse
|
756
|
Thottasseri AA, Rajendran V, Ramesh D, Tom AA, Thomas RR, Ray S, Gopan G, Mani M, Kannan T. Targeting Blood-Stage Malaria: Design, Synthesis, Characterization, In Vitro, and In Silico Evaluation of Pyrrolidinodiazenyl Chalcones. Chem Biol Drug Des 2025; 105:e70081. [PMID: 40070234 DOI: 10.1111/cbdd.70081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/25/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025]
Abstract
Malaria is a pervasive and deadly threat to the global population, and the resources available to treat this disease are limited. There is widespread clinical resistance to the most commonly prescribed antimalarial drugs. To address this issue, we synthesized a range of 4'-pyrrolidinodiazenyl chalcones using a covalent bitherapy approach to study their potential antimalarial properties. We examined the structure-activity relationships of these compounds, which could explain their antimalarial activities. The in vitro blood stage antimalarial activity of the compounds was evaluated against the mixed-blood stage culture (ring, trophozoites and schizonts) of Plasmodium falciparum 3D7, and the 50% inhibitory concentrations (IC50s) ranged from 3.3 to 22.2 μg/mL after 48 h of exposure. Compounds 11, 19, and 22 displayed pronounced IC50 values of 7.6 μg/mL, 6.4 μg/mL, and 3.3 μg/mL, respectively. The in vitro cytotoxicity of the active compounds was evaluated on human-derived Mo7e cells and murine-derived BA/F3 cells. Compounds 11 and 19 were found to be noncytotoxic (> 40 μg/mL), whereas compound 22 displayed cytotoxicity at higher concentrations. Moreover, these compounds exerted negligible hemolytic effects on human RBCs at their active concentrations. Molecular docking of these compounds revealed good hydrophobic and hydrogen bonding interactions with the binding sites of Plasmodium falciparum-dihydrofolate reductase, providing a rationale for their antimalarial activity, which is consistent with the in vitro results.
Collapse
Affiliation(s)
| | - Vinoth Rajendran
- Department of Microbiology, Pondicherry University, Kalapet, Puducherry, India
| | - Deepthi Ramesh
- Department of Chemistry, Pondicherry University, Kalapet, Puducherry, India
| | - Anju Agnes Tom
- Department of Chemistry, Pondicherry University, Kalapet, Puducherry, India
| | - Roshiny Roy Thomas
- Department of Chemistry, Pondicherry University, Kalapet, Puducherry, India
| | - Sreetama Ray
- Department of Chemistry, Pondicherry University, Kalapet, Puducherry, India
| | - Gopika Gopan
- Department of Microbiology, Pondicherry University, Kalapet, Puducherry, India
| | - Maheswaran Mani
- Department of Microbiology, Pondicherry University, Kalapet, Puducherry, India
| | | |
Collapse
|
757
|
Batin Rahaman SK, Halder S, Roy KK, Halder PK, Debnath U, Jana K. Discovery of New 4-Aminoquinoline-Thiazolidinone Hybrid Analogs as Antiproliferative Agents Inhibiting TLR4-LPS-Mediated Migration in Triple-Negative Breast Cancer Cells. Chem Biol Drug Des 2025; 105:e70089. [PMID: 40125628 DOI: 10.1111/cbdd.70089] [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: 11/23/2024] [Revised: 01/15/2025] [Accepted: 01/31/2025] [Indexed: 03/25/2025]
Abstract
The Toll-like receptor 4 (TLR4) signaling pathway plays a leading role in triggering proinflammatory responses by targeting lipopolysaccharide (LPS) molecules from different bacteria. Meanwhile, it is also expressed at higher levels in breast cancer cells than in normal breast tissue. After LPS binding, it initiates downstream signaling pathways that promote inflammation and cell apoptosis. Thus, targeting TLR4-LPS presents a promising dual therapeutic strategy for breast cancer treatment by not only inhibiting tumor growth but also reducing inflammation within the tumor microenvironment. To achieve this, the discovery of a new antiinflammatory agent is needed to reduce LPS-mediated cancer cell proliferation and migration. In this study, a series of 4-aminoquinoline-thiazolidinone hybrid analogs (4a-m) have been synthesized to explore their antiinflammatory as well as anticancer activity to find a new lead. Among them, 4e revealed the most promising antiinflammatory (IC50 = 2.38 ± 0.77 μM) as well as anticancer activity (IC50 = 3.26 ± 1.06 μM) in RAW 267.7 cell line and triple-negative breast cancer (TNBC) cell line, respectively. Further structure-activity relationship study followed by MD simulation analysis was carried out to identify probable binding residues of TLR4 which may play a significant role in developing antiinflammatory activity for promoting cell apoptosis in cancer cells.
Collapse
Affiliation(s)
- S K Batin Rahaman
- Department of Pharmaceutical Science, School of Health Sciences and Technology, UPES, Dehradun, India
| | - Satyajit Halder
- Department of Molecular Medicine, Bose Institute, Kolkata, India
| | - Kuldeep K Roy
- Department of Pharmaceutical Science, School of Health Sciences and Technology, UPES, Dehradun, India
| | - Pallab K Halder
- School of Natural Product Studies, Jadavpur University, Kolkata, India
| | - Utsab Debnath
- Department of Pharmaceutical Science, School of Health Sciences and Technology, UPES, Dehradun, India
| | - Kuladip Jana
- Department of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
758
|
Diaconu D, Savu M, Ciobanu C, Mangalagiu V, Mangalagiu II. Current strategies in design and synthesis of antifungals hybrid and chimeric diazine derivatives. Bioorg Med Chem 2025; 119:118069. [PMID: 39818112 DOI: 10.1016/j.bmc.2025.118069] [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: 10/21/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025]
Abstract
In the last decades fungal infections became a major threat to human health having an unacceptably occurrence, a high rate of mortality and the number of patients at risk for these infections continue to increase every year. An effective, modern and very useful strategy in antifungal therapy is represented by the use of chimeric and hybrid drugs, most of them being with azaheterocycle skeleton. In this review, we present an overview from the last five years of the most representative achievements in the field of chimeric and hybrid diazine derivatives with antifungal properties. Within this work we emphasize the most relevant data concerning the synthesis, design, Structure Activity Relationships (SAR) correlations and antifungal activity of the main classes of diazine: 1,2-diazine (pyridazine), 1,3-diazine (pyrimidine), 1,4-diazine (pyrazine) and their fused derivatives.
Collapse
Affiliation(s)
- Dumitrela Diaconu
- Alexandru Ioan Cuza University of Iasi, Faculty of Chemistry, Bd. Carol 11, 700506 Iasi, Romania; Alexandru Ioan Cuza University of Iasi, Institute of Interdisciplinary Research, RECENT-AIR Center, Bd. Carol 11, 700506 Iasi, Romania
| | - Marius Savu
- Alexandru Ioan Cuza University of Iasi, Faculty of Chemistry, Bd. Carol 11, 700506 Iasi, Romania
| | - Catalina Ciobanu
- Alexandru Ioan Cuza University of Iasi, Institute of Interdisciplinary Research, CERNESIM Center, Bd. Carol 11, 700506 Iasi, Romania
| | - Violeta Mangalagiu
- Alexandru Ioan Cuza University of Iasi, Institute of Interdisciplinary Research, CERNESIM Center, Bd. Carol 11, 700506 Iasi, Romania; Stefan Cel Mare University of Suceava, Faculty of Food Engineering, 13 Universitatii Str., 720229 Suceava, Romania
| | - Ionel I Mangalagiu
- Alexandru Ioan Cuza University of Iasi, Faculty of Chemistry, Bd. Carol 11, 700506 Iasi, Romania.
| |
Collapse
|
759
|
Yu S, Hu Z, Xu X, Liang X, Shen J, Liu M, Lin M, Chen H, Marti J, Tao SC, Xu Z. c-di-GMP inhibits rRNA methylation and impairs ribosome assembly in the presence of kanamycin. EMBO Rep 2025; 26:1367-1384. [PMID: 39870966 PMCID: PMC11894153 DOI: 10.1038/s44319-025-00377-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/20/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Cyclic diguanosine monophosphate (c-di-GMP) is a ubiquitous bacterial secondary messenger with diverse functions. A previous Escherichia coli proteome microarray identified that c-di-GMP binds to the 23S rRNA methyltransferases RlmI and RlmE. Here we show that c-di-GMP inhibits RlmI activity in rRNA methylation assays, and that it modulates ribosome assembly in the presence of kanamycin. Molecular dynamics simulation and mutagenesis studies reveal that c-di-GMP binds to RlmI at residues R64, R103, G114, and K201. Structural simulations indicate that c-di-GMP quenches RlmI activity by inducing the closure of the catalytic pocket. We also show that c-di-GMP promotes antibiotic tolerance through RlmI. Binding and methylation assays indicate that the inhibitory effect of c-di-GMP on RlmI is conserved across various pathogenic bacteria. Our data suggest an unexpected role for c-di-GMP in regulating ribosome assembly under stress through the inhibition of rRNA methyltransferases.
Collapse
Affiliation(s)
- Siqi Yu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Laboratory of Scientific Research, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Zheyao Hu
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, Barcelona, Catalonia, Spain
| | - Xiaoting Xu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
- Department of Endoscopy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaoran Liang
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Laboratory of Scientific Research, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Jiayi Shen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Laboratory of Scientific Research, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Min Liu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Laboratory of Scientific Research, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Mingxi Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
- Laboratory of Scientific Research, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Hong Chen
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Jordi Marti
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, Barcelona, Catalonia, Spain.
| | - Sheng-Ce Tao
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
| | - Zhaowei Xu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
- Laboratory of Scientific Research, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
760
|
Liang LJ, He B, Liang Y, Li YZ, Li ZM, Liu RB, Zhu TT, Luo Y, Lian XL, Zhao DH, Sun J, Ren H, Liao XP. Alisol A 24-Acetate combats Methicillin-Resistant Staphylococcus aureus infection by targeting the mevalonate biosynthesis. Biochem Pharmacol 2025; 233:116766. [PMID: 39894304 DOI: 10.1016/j.bcp.2025.116766] [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: 10/07/2024] [Revised: 12/31/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Infections caused by Methicillin-resistant Staphylococcus aureus (MRSA) have emerged as one of the most pressing global public health challenges. In concert with global rise of antimicrobial resistance at alarming rate, there is an urgent need for alternative strategies to combat MRSA. Here, the high throughput screening indicated that the Alisol A 24-acetate (AA) effectively inhibits the mevalonate (MVA) synthesis in MRSA. The mechanistic analysis revealed that AA competitively inhibits the 3-hydroxy-3-methyl glutaryl coenzyme A reductase (HMGR) protein to blockade the MVA pathway, thereby disrupting the bacterial membrane integrity and functions. Further investigations showed that this disruption consequently restores the β-lactam susceptibility in MRSA by retarding the expression of PBP2a protein and dampens the virulence of MRSA by reducing the exotoxins secretion. In addition to the effect on MRSA, AA has been found to exert host-acting activity to reduce the MRSA-induced inflammation. The promising anti-MRSA activity of AA was further confirmed in vivo. Collectively, the current study highlighted the potential of AA as a proposing drug for combating MRSA and emphasize the MVA pathway as an ideal therapeutic target for MRSA treatment.
Collapse
Affiliation(s)
- Li-Jie Liang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Bing He
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Yin Liang
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Yu-Ze Li
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Ze-Miao Li
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Rui-Bing Liu
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Ting-Ting Zhu
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Yang Luo
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Xin-Lei Lian
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Dong-Hao Zhao
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Hao Ren
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Xiao-Ping Liao
- Guangdong Laboratory for Lingnan Modern Agriculture, National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
761
|
Lundquist KP, Romeo I, Puglielli RB, Pestalozzi M, Gram ML, Hudson ES, Levi O, Arava YS, Gotfredsen CH, Clausen MH. Design, synthesis, and screening of an RNA optimized fluorinated fragment library. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100215. [PMID: 39828142 DOI: 10.1016/j.slasd.2025.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 12/30/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Fragment-based screening is an efficient method for early-stage drug discovery. In this study, we aimed to create a fragment library optimized for producing high hit rates against RNA targets. RNA has historically been an underexplored target, but recent research suggests potential for optimizing small molecule libraries for RNA binding. We extended this concept to fragment libraries to produce an RNA optimized fluorinated fragment library. We then screened this library, alongside two non-RNA optimized fragment libraries, against three RNA targets: the human cytoplasmic A-site and the S. cerevisiae tRNAAsp anticodon stem loop with and without nucleobase modifications. The screens yielded 24, 31, and 20 hits against the respective targets. Importantly, statistical analysis confirmed a significant overrepresentation of hits in our RNA optimized library. Based on these findings, we propose guidelines for developing RNA optimized fragment libraries. We hope the guidelines will help expediting fragment-based ligand discovery for RNA targets and contribute to presenting RNA as a promising target in drug discovery.
Collapse
Affiliation(s)
- Kasper P Lundquist
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Isabella Romeo
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | | | - Maëlle Pestalozzi
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Marie L Gram
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Emily S Hudson
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark
| | - Ofri Levi
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa 3200001, Israel
| | - Yoav S Arava
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa 3200001, Israel
| | - Charlotte H Gotfredsen
- NMR Center • DTU, DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark.
| | - Mads H Clausen
- DTU Chemistry, Technical University of Denmark, Kgs. Lyngby 2800, Denmark.
| |
Collapse
|
762
|
Li YH, Lin YK, Cai JF, Zou ZK, Zhao PL. A perspective on the application of macrocyclic design strategies in antitumor drugs. Bioorg Chem 2025; 156:108190. [PMID: 39855108 DOI: 10.1016/j.bioorg.2025.108190] [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: 10/23/2024] [Revised: 11/27/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
The macrocyclization of inhibitors has become gradually favored as a new approach for drug design in the field of anticancer agents, since the recent approvals of lorlatinib, pacritinib, and repotrectinib, and feasibility of macrocyclic modification to improve inhibitor drug-like properties has also been confirmed. Macrocycles are receiving increasing attention due to their enhanced binding affinity, target selectivity, and pharmacokinetic properties through conformational constraints. Therefore, this review summarizes various strategies for improving drug-like properties in macrocyclization and structural optimization, and reveals that macrocyclization is a new favorable strategy for drug design, aiming to provide insights for the drug discovery in different targets.
Collapse
Affiliation(s)
- Yan-Hong Li
- Guangdong Jiangmen Chinese Medicine College, Jiangmen 529000 PR China
| | - Yu-Kang Lin
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515 PR China
| | - Jian-Fan Cai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515 PR China
| | - Zhong-Kai Zou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515 PR China
| | - Pei-Liang Zhao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515 PR China.
| |
Collapse
|
763
|
Valapil DG, Devabattula G, Barahdia AS, Godugu C, Shankaraiah N. Development of 3-indolyl substituted phenyl pyrazolo-carboxamide hybrids as potential type II VEGFR-2 inhibitors and in vitro cytotoxicity studies. Bioorg Med Chem Lett 2025; 117:130070. [PMID: 39674381 DOI: 10.1016/j.bmcl.2024.130070] [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: 06/24/2024] [Revised: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
The progression of tumors is intricately linked to angiogenesis, the formation of new blood vessels, driven primarily by the release of growth factors such as Vascular Endothelial Growth Factor (VEGF). Targeting VEGF signaling through its receptor kinase (VEGFR-2) has emerged as a promising anti-angiogenic strategy for cancer therapy. In this study, we designed and synthesized a series of novel chemical entities based on 3-indolyl substituted phenyl pyrazole-carboxamides through docking studies upon considering the structure of sorafenib and its pattern of type II inhibition of VEGFR-2. Among the synthesized hybrids, 7b was able to significantly inhibit the growth of cancer cell lines, specifically against MCF-7 at 2.12 ± 0.19 μM. Compound 7b also efficiently inhibited VEGFR-2 kinase at a concentration of 2.83 ± 0.86 μM during the in vitro studies. Mechanistic studies revealed that 7b induced apoptosis evidenced by AO/EB, DAPI, and DCFDA staining, and its impact on the migratory ability of the cancer cells were also studied. These findings highlight the potential of 7b as a lead candidate for further development of anti-angiogenic therapies targeting VEGFR-2.
Collapse
Affiliation(s)
- Durgesh Gurukkala Valapil
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Geetanjali Devabattula
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Aman Singh Barahdia
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Chandraiah Godugu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| |
Collapse
|
764
|
Bedewy WA, Mulawka JW, Adler MJ. Classifying covalent protein binders by their targeted binding site. Bioorg Med Chem Lett 2025; 117:130067. [PMID: 39667507 DOI: 10.1016/j.bmcl.2024.130067] [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: 10/04/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Covalent protein targeting represents a powerful tool for protein characterization, identification, and activity modulation. The safety of covalent therapeutics was questioned for many years due to the possibility of off-target binding and subsequent potential toxicity. Researchers have recently, however, demonstrated many covalent binders as safe, potent, and long-acting therapeutics. Moreover, they have achieved selective targeting among proteins with high structural similarities, overcome mutation-induced resistance, and obtained higher potency compared to non-covalent binders. In this review, we highlight the different classes of binding sites on a target protein that could be addressed by a covalent binder. Upon folding, proteins generate various concavities available for covalent modifications. Selective targeting to a specific site is driven by differences in the geometry and physicochemical properties of the binding pocket residues as well as the geometry and reactivity of the covalent modifier "warhead". According to the warhead reactivity and the nature of the binding region, covalent binders can alter or lock a targeted protein conformation and inhibit or enhance its activity. We survey these various modification sites using case studies of recently discovered covalent binders, bringing to the fore the versatile application of covalent protein binders with respect to drug discovery approaches.
Collapse
Affiliation(s)
- Walaa A Bedewy
- Department of Chemistry & Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Egypt.
| | - John W Mulawka
- Department of Chemistry & Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| | - Marc J Adler
- Department of Chemistry & Biology, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada.
| |
Collapse
|
765
|
Elseginy SA. Exploring binding mechanisms of omicron spike protein with dolutegravir and etravirine by molecular dynamics simulation, principal component analysis, and free binding energy calculations. J Biomol Struct Dyn 2025; 43:2059-2072. [PMID: 38084778 DOI: 10.1080/07391102.2023.2293278] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/26/2023] [Indexed: 02/01/2025]
Abstract
The COVID-19 pandemic was caused by the SARS-CoV-2 virus, frequent mutations occurred to the wild-type virus resulting in evolved new variants. The WHO classified the new variants as 'Variants of Concern'. SARS-CoV-2 omicron evolved as the dominating variant at the end of 2021. Dolutegravir and etravirine were identified as inhibitors of SARS-CoV-2 entry to host cells in Omicron variants. In this study, combined in silico methods such as molecular docking, molecular dynamics, Principal component analysis, binding-free energy calculations, and Per Residues calculations were applied to investigate the mechanism of the bindings of the two inhibitors. The molecular dynamics results revealed the stability of dolutegravir-spike and etravirine-spike complexes in a similar manner to apo-protein. Dolutegravir and etravirine formed H-bonds and salt bridges with Omicron spike protein. The 2,4-difluoro phenyl moiety of dolutegravir plays an important role in binding the ligand. The binding mode and interactions of the two compounds indicated that Arg403, Tyr449, Tyr453, Arg493, Ser496, Arg498, Thr500, Tyr501, Gln502 and His505 are the key residues. The Principal Component Analyses suggested that no significant conformational changes happened for the two complexes during the simulations. Binding-free energy calculations showed that van der Waals interactions were the most important interactions for ligands' binding. Per-residue free energy decomposition revealed residues Arg493, Arg498, and Tyr501 contributed to the binding of the ligands through H-bonds and salt bridges formation while His505 contributed to H-bonds and Pi-Pi stacking and Phe497 contributed to hydrophobic interactions between ligands and Omicron spike protein.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Samia A Elseginy
- Green Chemistry Department, Chemical Industries Research Division, National Research Centre, Giza, Egypt
| |
Collapse
|
766
|
Pan B, Lin C, Lee GA, Lin H. Targeting SETDB1 in cancer and immune regulation: Potential therapeutic strategies in cancer. Kaohsiung J Med Sci 2025; 41:e12933. [PMID: 39764697 PMCID: PMC11924802 DOI: 10.1002/kjm2.12933] [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: 12/05/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 03/12/2025] Open
Abstract
SET domain bifurcated histone lysine methyltransferase 1 (SETDB1/ESET), a pivotal H3K9 methyltransferase, has been extensively studied since its discovery over two decades ago. SETDB1 plays critical roles in immune regulation, including B cell maturation, T-cell activity modulation, and endogenous retrovirus (ERV) silencing. While essential for normal immune cell function, SETDB1 overexpression in cancer cells disrupts immune responses by suppressing tumor immunogenicity and facilitating immune evasion. This is achieved through the repression of anti-tumor immune cell production, ERV silencing, and interference with the type I interferon pathway leading to inhibiting immune checkpoint blockade (ICB) efficacy. Beyond its immunological implications, SETDB1 overexpression fosters tumor growth and metastasis via transcriptional silencing of tumor suppressor genes through histone regulation and activating oncogenic signaling by non-histone regulation. These multifaceted roles make SETDB1 an attractive epigenetic target for novel cancer therapies. This review explores SETDB1's dual function in immune regulation and tumor progression, emphasizing its potential in the development of innovative cancer treatments targeting epigenetic dysregulation and oncogenic signaling.
Collapse
Affiliation(s)
- Bo‐Syong Pan
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Cheng‐Yu Lin
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Gilbert Aaron Lee
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Hui‐Kuan Lin
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
| |
Collapse
|
767
|
Thanh ND, Toan VN, Trang VM. D-glucose-conjugated thioureas containing 2-aminopyrimidine as potential multitarget inhibitors for type 2 diabetes mellitus: Synthesis and biological activity study. Comput Biol Med 2025; 186:109715. [PMID: 39862470 DOI: 10.1016/j.compbiomed.2025.109715] [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: 09/25/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
α-d-Glucose-conjugated thioureas 8a-w of substituted 4,6-diaryl-2-aminopyrimindines were designed, synthesized, and screened for their antidiabetic inhibitory activity. The thioureas with the strongest potential inhibitory activity included 8f (IC50 = 11.32 ± 0.34 μM for α-amylase), 8g (IC50 = 10.35 ± 0.88 μM for α-glucosidase), 8e (IC50 = 2.53 ± 0.03 nM for DPP-4), and 8c (IC50 = 3.93 ± 0.03 nM for PTP1B). The inhibitors 8g, 8e, and 8c were competitive α-glucosidase, non-competitive DDP-4, and non-competitive PTP1B inhibitors, respectively. In addition, compounds 8a, 8c, 8e, 8f, 8g, 8h, and 8j were noncytotoxic for 3T3 cell line. Induced fit docking study showed the key active interactions of each ligand with residues in the active site of each of these enzymes. Molecular dynamics simulation study on the representative complexes 8f/4W93 and 8e/3W2T in enzymes 4W93 and 3W2T, respectively, displayed the bioactive interactions between the residues and the corresponding potent inhibitor in the active site. Some of the various effects of the electron-donating and electron-withdrawing substituents on benzene of pyrimidine ring to inhibitory activities against enzymes related to T2DM were discussed. The calculations based on MM-GBSA showed the effects of the solvation to the active binding of the specific ligand in the active pocket of an enzyme.
Collapse
Affiliation(s)
- Nguyen Dinh Thanh
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi), 19 Le Thanh Tong, Hoan Kiem, Ha Noi, Viet Nam.
| | - Vu Ngoc Toan
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi), 19 Le Thanh Tong, Hoan Kiem, Ha Noi, Viet Nam; Institute of New Technology, Academy of Military Science and Technology, Ministry of Defence, 17 Hoang Sam, Cau Giay, Ha Noi, Viet Nam
| | - Vu Minh Trang
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi), 19 Le Thanh Tong, Hoan Kiem, Ha Noi, Viet Nam; VNU University of Education, Vietnam National University, Hanoi, 144 Xuan Thuy, Cau Giay, Ha Noi, Viet Nam
| |
Collapse
|
768
|
Fantini J, Azzaz F, Di Scala C, Aulas A, Chahinian H, Yahi N. Conformationally adaptive therapeutic peptides for diseases caused by intrinsically disordered proteins (IDPs). New paradigm for drug discovery: Target the target, not the arrow. Pharmacol Ther 2025; 267:108797. [PMID: 39828029 DOI: 10.1016/j.pharmthera.2025.108797] [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: 09/01/2024] [Revised: 11/28/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
The traditional model of protein structure determined by the amino acid sequence is today seriously challenged by the fact that approximately half of the human proteome is made up of proteins that do not have a stable 3D structure, either partially or in totality. These proteins, called intrinsically disordered proteins (IDPs), are involved in numerous physiological functions and are associated with severe pathologies, e.g. Alzheimer, Parkinson, Creutzfeldt-Jakob, amyotrophic lateral sclerosis (ALS), and type 2 diabetes. Targeting these proteins is challenging for two reasons: i) we need to preserve their physiological functions, and ii) drug design by molecular docking is not possible due to the lack of reliable starting conditions. Faced with this challenge, the solutions proposed by artificial intelligence (AI) such as AlphaFold are clearly unsuitable. Instead, we suggest an innovative approach consisting of mimicking, in short synthetic peptides, the conformational flexibility of IDPs. These peptides, which we call adaptive peptides, are derived from the domains of IDPs that become structured after interacting with a ligand. Adaptive peptides are designed with the aim of selectively antagonizing the harmful effects of IDPs, without targeting them directly but through selected ligands, without affecting their physiological properties. This "target the target, not the arrow" strategy is promised to open a new route to drug discovery for currently undruggable proteins.
Collapse
Affiliation(s)
- Jacques Fantini
- Aix-Marseille University, INSERM UA 16, Faculty of Medicine, 13015 Marseille, France.
| | - Fodil Azzaz
- Aix-Marseille University, INSERM UA 16, Faculty of Medicine, 13015 Marseille, France
| | - Coralie Di Scala
- Neuroscience Center-HiLIFE, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Anaïs Aulas
- Neuroscience Center-HiLIFE, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Henri Chahinian
- Aix-Marseille University, INSERM UA 16, Faculty of Medicine, 13015 Marseille, France
| | - Nouara Yahi
- Aix-Marseille University, INSERM UA 16, Faculty of Medicine, 13015 Marseille, France
| |
Collapse
|
769
|
Xu Z, Liu J, Zhuang Y. The anti-Acinetobacter baumannii therapeutic potential of azole hybrids: A mini-review. Arch Pharm (Weinheim) 2025; 358:e2400592. [PMID: 40040257 DOI: 10.1002/ardp.202400592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 12/19/2024] [Accepted: 02/11/2025] [Indexed: 03/06/2025]
Abstract
Acinetobacter baumannii is one of the major causes of severe hospital- and community-acquired infections, posing a significant threat to human lives. A. baumannii has already generated resistance to almost all of the currently available antibiotics, but no new class of antibacterials have been launched for the treatment of infections caused by A. baumannii in the last half century, creating an urgent need to develop novel antibacterials. Azoles as a broad class of five-membered nitrogen-containing aromatic heterocycles are privileged pharmacophores widely found in pharmaceuticals. Azoles could target on diverse enzymes, proteins, and receptors in A. baumannii via various noncovalent interactions. Particularly, azole hybrids have potential advantages in increasing therapeutic efficacy and circumventing drug resistance, representing useful scaffolds for the discovery of novel anti-A. baumannii agents. This review outlines the current scenario of the antibacterial therapeutic potential of azole hybrids against A. baumannii, developed from 2020 onwards, aiming to provide potential candidates for further preclinical/clinical evaluations and facilitate the rational design of more effective candidates.
Collapse
Affiliation(s)
- Zhi Xu
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, People's Republic of China
| | - Junna Liu
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, People's Republic of China
| | - Yafei Zhuang
- Huanghuai University Industry Innovation & Research and Development Institute, Huanghuai University, Zhumadian, People's Republic of China
| |
Collapse
|
770
|
Takjoo R, Wilson DT, Le Quilliec J, Schmidt CA, Zhao G, Liddell MJ, Shaikh NY, Sunagar K, Loukas A, Smout MJ, Daly NL. Structural analysis of an Asterias rubens peptide indicates the presence of a disulfide-directed β-hairpin fold. FEBS Open Bio 2025; 15:415-426. [PMID: 39561265 PMCID: PMC11891777 DOI: 10.1002/2211-5463.13931] [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: 05/24/2024] [Revised: 10/19/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
Sea stars are an abundant group of marine invertebrates that display remarkably robust regenerative capabilities throughout all life stages. Numerous proteins and peptides have been identified in a proteome study on the coelomic fluid (biofluid) of the common sea star Asterias rubens, which appear to be involved with the wound-healing response in the organism. However, the three-dimensional structure and function of several of these injury-responsive peptides, including the peptide KASH2, are yet to be investigated. Here, we show that the KASH2 peptide adopts a disulfide-directed β-hairpin fold (DDH). The DDH motif appears to be evolutionarily related to the inhibitor cystine knot motif, which is one of the most widespread disulfide-rich peptide folds. The DDH motif was originally thought to be restricted to arachnids, but our study suggests that as a result of convergent evolution it could also have originated in sea stars. Although the widely conserved DDH fold has potential cross-phyla wound-healing capacity, we have shown that KASH2 does not enhance the proliferation of human fibroblasts, a simple method for wound-healing re-epithelialisation screening. Therefore, additional research is necessary to determine the role of KASH2 in the sea stars.
Collapse
Affiliation(s)
- Rozita Takjoo
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - David T. Wilson
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - Justine Le Quilliec
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
- Univ Brest, École Supérieure d'Ingénieurs en Agroalimentaire de Bretagne atlantiquePlouzanéFrance
| | - Casey A. Schmidt
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - Guangzu Zhao
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | | | - Naeem Y. Shaikh
- Evolutionary Venomics Lab, Centre for Ecological SciencesIndian Institute of ScienceBangaloreIndia
| | - Kartik Sunagar
- Evolutionary Venomics Lab, Centre for Ecological SciencesIndian Institute of ScienceBangaloreIndia
| | - Alex Loukas
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - Michael J. Smout
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| | - Norelle L. Daly
- Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsAustralia
| |
Collapse
|
771
|
Jin Y, Qi X, Yu X, Cheng X, Chen B, Wu M, Zhang J, Yin H, Lu Y, Zhou Y, Pang A, Lin Y, Jiang L, Shi Q, Geng S, Zhou Y, Yao X, Li L, Duan H, Che J, Cao J, He Q, Dong X. Discovery of a potential hematologic malignancies therapy: Selective and potent HDAC7 PROTAC degrader targeting non-enzymatic function. Acta Pharm Sin B 2025; 15:1659-1679. [PMID: 40370550 PMCID: PMC12069251 DOI: 10.1016/j.apsb.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/10/2024] [Accepted: 12/18/2024] [Indexed: 05/16/2025] Open
Abstract
HDAC7, a member of class IIa HDACs, plays a pivotal regulatory role in tumor, immune, fibrosis, and angiogenesis, rendering it a potential therapeutic target. Nevertheless, due to the high similarity in the enzyme active sites of class IIa HDACs, inhibitors encounter challenges in discerning differences among them. Furthermore, the substitution of key residue in the active pocket of class IIa HDACs renders them pseudo-enzymes, leading to a limited impact of enzymatic inhibitors on their function. In this study, proteolysis targeting chimera (PROTAC) technology was employed to develop HDAC7 drugs. We developed an exceedingly selective HDAC7 PROTAC degrader B14 which showcased superior inhibitory effects on cell proliferation compared to TMP269 in various diffuse large B cell lymphoma (DLBCL) and acute myeloid leukemia (AML) cells. Subsequent investigations unveiled that B14 disrupts BCL6 forming a transcriptional inhibition complex by degrading HDAC7, thereby exerting proliferative inhibition in DLBCL. Our study broadened the understanding of the non-enzymatic functions of HDAC7 and underscored the importance of HDAC7 in the treatment of hematologic malignancies, particularly in DLBCL and AML.
Collapse
Affiliation(s)
- Yuheng Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xuxin Qi
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Xiaoli Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xirui Cheng
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Boya Chen
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Mingfei Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jingyu Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hao Yin
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Yang Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yihui Zhou
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Ao Pang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yushen Lin
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Li Jiang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Qiuqiu Shi
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shuangshuang Geng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yubo Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Guangdong 528400, China
| | - Xiaojun Yao
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China
| | - Linjie Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Haiting Duan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jinxin Che
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ji Cao
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Cancer Center, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou 310000, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou 310020, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences and Cancer Center, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou 310000, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Xiaowu Dong
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
772
|
Chen B, Liu S, Xia H, Li X, Zhang Y. Computer-Aided Drug Design in Research on Chinese Materia Medica: Methods, Applications, Advantages, and Challenges. Pharmaceutics 2025; 17:315. [PMID: 40142979 PMCID: PMC11945071 DOI: 10.3390/pharmaceutics17030315] [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: 02/12/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Chinese materia medica (CMM) refers to the medicinal substances used in traditional Chinese medicine. In recent years, CMM has become globally prevalent, and scientific research on CMM has increasingly garnered attention. Computer-aided drug design (CADD) has been employed in Western medicine research for many years, contributing significantly to its progress. However, the role of CADD in CMM research has not been systematically reviewed. This review briefly introduces CADD methods in CMM research from the perspectives of computational chemistry (including quantum chemistry, molecular mechanics, and quantum mechanics/molecular mechanics) and informatics (including cheminformatics, bioinformatics, and data mining). Then, it provides an exhaustive discussion of the applications of these CADD methods in CMM research through rich cases. Finally, the review outlines the advantages and challenges of CADD in CMM research. In conclusion, despite the current challenges, CADD still offers unique advantages over traditional experiments. With the development of the CMM industry and computer science, especially driven by artificial intelligence, CADD is poised to play an increasingly pivotal role in advancing CMM research.
Collapse
Affiliation(s)
- Ban Chen
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; (B.C.); (S.L.); (H.X.)
| | - Shuangshuang Liu
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; (B.C.); (S.L.); (H.X.)
| | - Huiyin Xia
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; (B.C.); (S.L.); (H.X.)
| | - Xican Li
- School of Chinese Herbal Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China;
| | - Yingqing Zhang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Centre of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan 430068, China; (B.C.); (S.L.); (H.X.)
| |
Collapse
|
773
|
Zhang N, Wang X, Zhu C, Noraddin A, Yu Y, Wang X, Shi Y, Chen K, Liu X, Zhang Y, Yu Z. Synthesis and biological evaluation of new dual APN/NEP inhibitors as potent analgesics. Bioorg Chem 2025; 156:108210. [PMID: 39879827 DOI: 10.1016/j.bioorg.2025.108210] [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: 10/22/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
An alternative approach for the management of acute and chronic pains involves prolonging the half-life of endogenous opiates, such as enkephalins that are released in response to nociceptive stimuli. This can be achieved through the inhibition of enzymatic pathways responsible for the hydrolysis of these peptides, particularly targeting Aminopeptidase N (APN) and Neutral Endopeptidase (NEP). In this study, we designed and synthesized a series of dual enkephalinase inhibitors (DENKIs) targeting both APN and NEP as novel analgesic treatments. Notably, SDUY812, SDUY816 and SDUY817 exhibited potent inhibition of APN activity with IC50 values of 0.38 µM, 0.68 µM and 0.29 µM, respectively, whereas their IC50 values against NEP were 6.9 µM, 6.9 µM and 7.4 µM, separately. In in-vivo antinociceptive assays, SDUY816 and SDUY817 demonstrated superior analgesic efficacy compared to Thiorphan and Bestatin in mice models of acute, inflammatory and neuropathic pains with jumping latencies exceeding 100 s and withdrawal thresholds more than 0.13 g. Moreover, the analgesic activity of these inhibitors was significantly diminished by a potent opioid antagonist, naloxone, indicating the contribution of opioid receptors to the robust analgesic properties of these newly developed DENKIs. In addition, SDUY816 and SDUY817 exerted the analgesic activity in a concentration- and time-dependent manner with SDUY816 possessing acceptable pharmacokinetic properties (t1/2 = 4.02 h and F = 27 %) and low toxicity. These findings provide alternative analgesic therapeutics that are potentially devoid of opioid-associated side effects.
Collapse
Affiliation(s)
- Naining Zhang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinyue Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Chengchun Zhu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Amina Noraddin
- Immagina Biotechnology S.r.l, Viale dell'Industria 47, 38057 Pergine Valsugana, TN, Italy; Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Yan Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiao Wang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Ying Shi
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Kai Chen
- Center for New Drug Evaluation, Shandong Academy of Pharmaceutical Sciences, Jinan 250000, China
| | - Xiaoyu Liu
- Center for New Drug Evaluation, Shandong Academy of Pharmaceutical Sciences, Jinan 250000, China
| | - Yan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Zhiyi Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
774
|
Yang J, Xie YF, Smith R, Ratté S, Prescott SA. Discordance between preclinical and clinical testing of Na V 1.7-selective inhibitors for pain. Pain 2025; 166:481-501. [PMID: 39928833 PMCID: PMC11808711 DOI: 10.1097/j.pain.0000000000003425] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 10/26/2024]
Abstract
ABSTRACT The voltage-gated sodium channel Na V 1.7 plays an important role in pain processing according to genetic data. Those data made Na V 1.7 a popular drug target, especially since its relatively selective expression in nociceptors promised pain relief without the adverse effects associated with broader sodium channel blockade. Despite encouraging preclinical data in rodents, Na V 1.7-selective inhibitors have not yet proven effective in clinical trials. Discrepancies between preclinical and clinical results should raise alarms. We reviewed preclinical and clinical reports on the analgesic efficacy of Na V 1.7-selective inhibitors and found critical differences in several factors. Putting aside species differences, most preclinical studies tested young male rodents with limited genetic variability, inconsistent with the clinical population. Inflammatory pain was the most common preclinical chronic pain model whereas nearly all clinical trials focused on neuropathic pain despite some evidence suggesting Na V 1.7 channels are not essential for neuropathic pain. Preclinical studies almost exclusively measured evoked pain whereas most clinical trials assessed average pain intensity without distinguishing between evoked and spontaneous pain. Nearly all preclinical studies gave a single dose of drug unlike the repeat dosing used clinically, thus precluding preclinical data from demonstrating whether tolerance or other slow processes occur. In summary, preclinical testing of Na V 1.7-selective inhibitors aligned poorly with clinical testing. Beyond issues that have already garnered widespread attention in the pain literature, our results highlight the treatment regimen and choice of pain model as areas for improvement.
Collapse
Affiliation(s)
- Jane Yang
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Russell Smith
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Steven A. Prescott
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
775
|
Singh AK, Kumar A, Singh H, Verma A, Kumar P. Refinement of MEK inhibitors. Future Med Chem 2025; 17:501-503. [PMID: 39910918 PMCID: PMC11901393 DOI: 10.1080/17568919.2025.2463311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Affiliation(s)
- Ankit Kumar Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
| | - Adarsh Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Harshwardhan Singh
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| |
Collapse
|
776
|
Martina MG, Rubini D, Radi M, Cagno V. Targeting PI4KB and Src/Abl host kinases as broad-spectrum antiviral strategy: Myth or real opportunity? Antiviral Res 2025; 235:106100. [PMID: 39922541 DOI: 10.1016/j.antiviral.2025.106100] [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: 12/16/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Viruses pose a continuous threat to human health. Limited treatment options exist for current viruses, and the risk of infections with newly emerging or re-emerging viruses is increasing. In a pandemic scenario, having a broad-spectrum antiviral to limit viral spread while developing specific antivirals and vaccines is crucial. Targeting host kinases represents a valuable strategy due to the higher barrier to resistance and the broad-spectrum activity it offers. While cells have redundant kinases for the same biological function, viruses rely on specific kinases for their replication cycle, enabling targeted antiviral action with limited toxicity. This review focuses on two extensively studied kinase targets: the lipid kinase phosphatidylinositol 4-kinase IIIβ (PI4KB) and the tyrosine kinase proteins Src and Abl. Compounds active against these targets are reviewed in terms of the viruses they inhibit, their mechanisms of action and their stage of development. While PI4KB inhibitors have reached clinical trials, those targeting Src and Abl remain largely in the preclinical phase. Nevertheless, opportunities exist to improve potency and further understand the specific roles of these kinases in the life cycle of multiple viruses.
Collapse
Affiliation(s)
- Maria Grazia Martina
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Daniele Rubini
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124, Parma, Italy.
| | - Valeria Cagno
- Institute of Microbiology, University Hospital of Lausanne, University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
777
|
Gaber AA, El-Morsy AM, Sherbiny FF, Bayoumi AH, El-Gamal KM, El-Adl K, Al-Karmalawy AA, Ezz Eldin RR, Saleh MA, Abulkhair HS. Pharmacophore-linked pyrazolo[3,4-d]pyrimidines as EGFR-TK inhibitors: Synthesis, anticancer evaluation, pharmacokinetics, and in silico mechanistic studies. Arch Pharm (Weinheim) 2025; 358:e2100258. [PMID: 34467546 DOI: 10.1002/ardp.202100258] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 02/05/2023]
Abstract
Targeting the epidermal growth factor receptors (EGFRs) with small inhibitor molecules has been validated as a potential therapeutic strategy in cancer therapy. Pyrazolo[3,4-d]pyrimidine is a versatile scaffold that has been exploited for developing potential anticancer agents. On the basis of fragment-based drug discovery, considering the essential pharmacophoric features of potent EGFR tyrosine kinase (TK) inhibitors, herein, we report the design and synthesis of new hybrid molecules of the pyrazolo[3,4-d]pyrimidine scaffold linked with diverse pharmacophoric fragments with reported anticancer potential. These fragments include hydrazone, indoline-2-one, phthalimide, thiourea, oxadiazole, pyrazole, and dihydropyrazole. The synthesized molecules were evaluated for their anticancer activity against the human breast cancer cell line, MCF-7. The obtained results revealed comparable antitumor activity with that of the reference drugs doxorubicin and toceranib. Docking studies were performed along with EGFR-TK and ADMET profiling studies. The results of the docking studies showed the ability of the designed compounds to interact with key residues of the EGFR-TK through a number of covalent and noncovalent interactions. The obtained activity of compound 25 (IC50 = 2.89 µM) suggested that it may serve as a lead for further optimization and drug development.
Collapse
Affiliation(s)
- Ahmed A Gaber
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
| | - Ahmed M El-Morsy
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
- Pharmaceutical Chemistry Department, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Farag F Sherbiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
- Department of Chemistry, Basic Science Center and Pharmaceutical Organic Chemistry College of Pharmaceutical Science & Drug Manufacturing, Misr University for Science and Technology (MUST), Al-Motamayez District, 6th of October City, Egypt
| | - Ashraf H Bayoumi
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
| | - Kamal M El-Gamal
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
| | - Khaled El-Adl
- Department of Medicinal Chemistry & Drug Design, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| | - Rogy R Ezz Eldin
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Marwa A Saleh
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Nasr City, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| |
Collapse
|
778
|
Huang Z, Zhang X, Li J, Zhang L, Shen Y, Wang R, Zhang Y, Mao Z. N-Heterocyclic functionalized chalcone derivatives as anti-inflammatory agents for atopic dermatitis treatment by inhibiting JAK1/STAT3 signaling pathway. Bioorg Chem 2025; 156:108200. [PMID: 39874907 DOI: 10.1016/j.bioorg.2025.108200] [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: 12/06/2024] [Revised: 12/29/2024] [Accepted: 01/19/2025] [Indexed: 01/30/2025]
Abstract
Atopic dermatitis (AD) is difficult to cure as a chronic inflammatory skin disease. In the present study, a series of N-heterocyclic functionalized chalcone derivatives have been prepared to investigate their in vitro and in vivo anti-inflammatory activities. The results indicated that many derivatives could effectively inhibit NO generation with low toxicity. In vivo studies revealed that 4f could improve the skin condition of AD-like mice, reduce inflammatory infiltration, inhibit the expressions of p-JAK1/JAK1 and p-STAT3/STAT3, and mitigate the excessive immune response on MC903-induced AD-like mice. The molecular docking study indicated that 4f had an obvious binding site with the target 4ehz and 6QHD. Therefore, these derivatives may be considered as potent agents for AD treatment by inhibiting JAK1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Zhengxiao Huang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xinjia Zhang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Jing Li
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Lijuan Zhang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yunhong Shen
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Ruirui Wang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yi Zhang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Zewei Mao
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
779
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
780
|
Kufa M, Finger V, Kovar O, Soukup O, Torruellas C, Roh J, Korabecny J. Revolutionizing tuberculosis treatment: Breakthroughs, challenges, and hope on the horizon. Acta Pharm Sin B 2025; 15:1311-1332. [PMID: 40370552 PMCID: PMC12069392 DOI: 10.1016/j.apsb.2025.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 05/16/2025] Open
Abstract
Tuberculosis (TB), an infectious disease caused by the bacterium Mycobacterium tuberculosis (Mtb), was responsible for the deaths of approximately 1.3 million people in 2022. In addition, 7.5 million new cases of TB have been reported. Present-day treatments require a daily dosing of a multiple-drug regimen for a minimum of six-month, but poor adherence and other factors often lead to treatment failure. Consequently, drug-resistant TB strains have become a growing concern, leading to more complex and expensive treatments. Promising drugs such as bedaquiline, delamanid, and pretomanid have been recently released, and 19 drug candidates are currently at different phases of clinical trials, addressing the problem of drug-resistant TB. Notwithstanding recent advances, the development of effective and safe drugs with novel mechanisms of action remains a challenge due to the unique nature of Mtb. Despite the persistent need for new treatments, TB research remains underfunded, highlighting the importance of collaborations between academia and the private sector in the advancement of anti-TB drug development. This review provides a perspective on the dynamic landscape of anti-TB drug discovery in recent years, offering hope for a more effective approach to combat this persistent global health threat.
Collapse
Affiliation(s)
- Martin Kufa
- Faculty of Pharmacy in Hradec Kralové, Charles University, Hradec Kralove 50003, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove 50003, Czech Republic
| | - Vladimir Finger
- Faculty of Pharmacy in Hradec Kralové, Charles University, Hradec Kralove 50003, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove 50003, Czech Republic
| | - Ondrej Kovar
- Faculty of Pharmacy in Hradec Kralové, Charles University, Hradec Kralove 50003, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove 50003, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove 50003, Czech Republic
| | | | - Jaroslav Roh
- Faculty of Pharmacy in Hradec Kralové, Charles University, Hradec Kralove 50003, Czech Republic
| | - Jan Korabecny
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove 50003, Czech Republic
| |
Collapse
|
781
|
Khedkar NR, Sindkhedkar M, Joseph A. Fragment-Based Drug Discovery: Small Fragments, Big Impact - Success Stories of Approved Oncology Therapeutics. Bioorg Chem 2025; 156:108197. [PMID: 39879825 DOI: 10.1016/j.bioorg.2025.108197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/01/2024] [Accepted: 01/18/2025] [Indexed: 01/31/2025]
Abstract
Fragment-Based Drug Discovery (FBDD) has revolutionized drug discovery by overcoming the challenges of traditional methods like combinatorial chemistry and high-throughput screening (HTS). Leveraging small, low-molecular-weight fragments, FBDD achieves higher hit rates, reduced screening costs, and faster development timelines for clinically relevant drug candidates. This review explores FBDD's core principles, innovative methodologies, and its success in targeting diverse protein classes, including previously "undruggable" targets. Key advancements in fragment libraries, screening techniques, and computational tools are discussed, along with the efficient progression from fragment hits to clinical drugs. Notably, we highlight FDA-approved fragment-derived drugs, including capivasertib, which has increased the total number of fragment-based oncology drugs to seven. As FBDD continues to evolve, its potential to address unmet therapeutic needs and drive the discovery of groundbreaking treatments across various disease areas becomes increasingly evident.
Collapse
Affiliation(s)
- Nilesh Raghunath Khedkar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal, Academy of Higher Education, Manipal, Karnataka 576104, India; Novel Drug Discovery & Development, Lupin Research Park, Lupin Ltd., Pune 412115, India; Research Scholar, Manipal Academy of Higher Education, India
| | - Milind Sindkhedkar
- Novel Drug Discovery & Development, Lupin Research Park, Lupin Ltd., Pune 412115, India.
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal, Academy of Higher Education, Manipal, Karnataka 576104, India
| |
Collapse
|
782
|
Zhou Y, Wang J, Sun Y, Cheng Y, Wu W. Non-Hydroxamate Inhibitors of IspC Enzyme in the MEP Pathway: Structural Insights and Drug Development Potential. Chem Biol Drug Des 2025; 105:e70086. [PMID: 40099748 DOI: 10.1111/cbdd.70086] [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: 11/13/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
1-Deoxy-D-xylulose-5-phosphate reductoisomerase (IspC) is a key enzyme in the MEP pathway, essential for many bacteria, human pathogens, and plants, thus being an attractive drug target. Fosmidomycin, a potent IspC inhibitor with hydroxamate metal-binding pharmacophores (MBPs), has entered clinical trials for malaria but is hampered by pharmacokinetic and toxicity issues of the hydroxamate fragment. This has led to increased interest in non-hydroxamate inhibitors. This review focuses on the crystal structure and active-site binding mode of IspC, and the structural types, inhibitory activities, and structure-activity relationships of non-hydroxamate IspC inhibitors. Early attempts to design such inhibitors involved direct removal or replacement of the hydroxamate MBPs, with varying results. Lipophilic inhibitors, bisubstrate inhibitors, and those developed for herbicidal applications have shown promise. However, challenges remain due to the sensitivity of the enzyme active site to ligand interactions. Future research could draw from other metalloenzyme studies to develop novel and efficient non-hydroxamate IspC inhibitors.
Collapse
Affiliation(s)
- Yaqing Zhou
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Jili Wang
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Yong Sun
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Yarui Cheng
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Wenhai Wu
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| |
Collapse
|
783
|
Khanka S, Rastogi SK, Singh KB, Sharma K, Parwez S, Siddiqi MI, Sinha AK, Kumar R, Singh D. Pym-18a, a novel pyrimidine derivative ameliorates glucocorticoid induced osteoblast apoptosis and promotes osteogenesis via autophagy and PINK 1/Parkin mediated mitophagy induction. Biochem Pharmacol 2025; 233:116751. [PMID: 39800267 DOI: 10.1016/j.bcp.2025.116751] [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: 08/14/2024] [Revised: 12/11/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is the most common type of secondary osteoporosis, marked by reduced bone density and impaired osteoblast function. Current treatments have serious side effects, highlighting the need for new drug candidates. Pyrimidine derivatives have been noted for their potential in suppressing osteoclastogenesis, but their effects on osteogenesis and GIOP remain underexplored. Our recent study identified a novel pyrimidine derivative, Pym-18a, which enhances osteoblast functions. In this study, Pym-18a was found to mitigate the detrimental effects of Dexamethasone (Dex) in osteoblast cells and in GIOP in Balb/C mice. Pretreatment with Pym-18a followed by Dex (100 µM) for 24 h restored osteoblast alkaline phosphatase activity and viability. Pym-18a reduced Dex-induced apoptosis and reactive oxygen species (ROS) generation at cellular and mitochondrial levels and preserved mitochondrial membrane potential. Dex impaired autophagy and mitophagy, however but Pym-18a pretreatment increased expression of autophagy markers (LC3II) and mitophagy markers (PINK1, Parkin, TOM20) while decreasing P62 expression. The osteogenic effects of Pym-18a were diminished in the presence of 3-MA (an autophagy inhibitor). In silico studies showed mTOR inhibition by Pym-18a, corroborated by its suppression of Dex-induced mTOR activation. In vivo, Pym-18a (10 mg/kg) significantly improved bone microarchitecture, trabecular connectivity, and strength, and corrected P1NP and CTX levels altered by Dex. Pym-18a also promoted autophagy, mitophagy, and suppressed mTOR activation in GIOP mice. Overall, Pym-18a mitigates detrimental effect of Dex by modulating autophagy and PINK/Parkin-mediated mitophagy through mTOR inhibition, suggesting it as a potential novel therapeutic option for GIOP.
Collapse
Affiliation(s)
- Sonu Khanka
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Sumit K Rastogi
- Division of Medicinal & Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Krishna Bhan Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Kriti Sharma
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Shahid Parwez
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Mohammad Imran Siddiqi
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Arun K Sinha
- Division of Medicinal & Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Ravindra Kumar
- Division of Medicinal & Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Divya Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
784
|
Khalaf HS, Abdel-Aziz MS, Radwan MAA, Sediek AA. Synthesis, Biological Evaluation, and Molecular Docking Studies of Indole-Based Heterocyclic Scaffolds as Potential Antibacterial Agents. Chem Biodivers 2025; 22:e202402325. [PMID: 39433506 DOI: 10.1002/cbdv.202402325] [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: 09/17/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 10/23/2024]
Abstract
Indole-based heterocyclic scaffolds have become increasingly important in medicinal chemistry due to their notable pharmacological and biological properties. Their role in the discovery and development of innovative drugs for treating various diseases highlights their value. This study aimed to synthesize C3-indole derivatives linked to various heterocyclic scaffolds, including thiophenes, thiazolidine-4-ones, and 1,3,4-thiadiazoles, via the reaction of ethylthioacetanilide 2 with different α-haloketones.The structures of the target compounds were established using 1H and 13C nuclear magnetic resonance spectroscopy, mass spectrometry, infrared spectroscopy, and elemental analysis. The synthesized compounds were tested for antimicrobial activity against different microbes: S. aureus ATCC 6538 (Gram-positive bacteria), E. coli ATCC 25933 (Gram-negative bacteria), C. albicans ATCC 10231 (yeast), and fungi (A. niger NRRL-A326). Thiophene 6a, thiazolidine-4-one 8, and compound 10d exhibited the highest antimicrobial activities. The molecular docking study showed that compounds 2, 4, 6a, and 6c had good binding energy and favorable binding modes of interactions with the DNA gyrase B enzymes (PDB: 3 U2D) and (PDB: 1S14). The results showed that the NH group of the indole in compounds 2 and 4, together with the nitrile group (CN), played an important role in inhibiting DNA gyrase B of S. aureus, PDB: 3 U2D. Furthermore, the NH of the indole ring, together with the ethylamino group of compound 2, was crucial in inhibiting DNA gyrase B of E. coli, PDB: 1S14. These findings may encourage researchers to develop more effective C3-indole derivatives in their search for antimicrobial drugs.
Collapse
Affiliation(s)
- Hemat S Khalaf
- Department of Photochemistry, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Mohamed S Abdel-Aziz
- Microbial Chemistry Department, Biotechnology Research Institute, National Research Centre, Dokki, Giza, Egypt
| | - Mohamed A A Radwan
- Applied Organic Chemistry Department, National Research Centre, Dokki, 12622, Egypt
| | - Ashraf A Sediek
- Chemical Industries Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| |
Collapse
|
785
|
Alasalvar C, Huang G, Bolling BW, Jantip PA, Pegg RB, Wong XK, Chang SK, Pelvan E, de Camargo AC, Mandalari G, Hossain A, Shahidi F. Upcycling commercial nut byproducts for food, nutraceutical, and pharmaceutical applications: A comprehensive review. Food Chem 2025; 467:142222. [PMID: 39626555 DOI: 10.1016/j.foodchem.2024.142222] [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: 08/09/2024] [Revised: 11/07/2024] [Accepted: 11/21/2024] [Indexed: 01/15/2025]
Abstract
This article presents a comprehensive overview of upcycling commercial nut byproducts (such as Brazil nut, cashew, hazelnut, macadamia, peanut (also known as a legume), pecan, pine nut, pistachio, and walnut) for food, nutraceutical, and pharmaceutical applications. Upcycling nut byproducts, namely husk/hull, hard shell, brown skin, defatted flour/meal/cake, pine cone, cashew nut shell liquid, cashew apple, walnut septum, and dreg/okara, has great potential, not only to reduce/minimise waste, but also to fit within the circular economy concept. Each byproduct has its own unique functional properties, which can bring significant value. These byproducts can be used as value-added ingredients to promote better health and well-being, due to their rich sources of diverse bioactive components/phytochemicals, polysaccharides, fibre, lignin, prebiotics, oils, proteins, bioactive peptides, minerals, and vitamins, among other components. This comprehensive review provides a basis for future research and development of product applications for nut byproducts. More studies are needed on novel product development to valorise nut byproducts.
Collapse
Affiliation(s)
| | | | - Bradley W Bolling
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Pornpat Aom Jantip
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Ronald B Pegg
- Department of Food Science & Technology, University of Georgia, Athens, GA, USA
| | - Xi Khai Wong
- School of Science, Monash University (Malaysia Campus), Jalan Lagoon Selatan, Bandar Sunway, 47500 Subang Jaya, Selangor, Malaysia
| | - Sui Kiat Chang
- Department of Allied Health Sciences, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia; Centre for Biomedical and Nutrition Research, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Ebru Pelvan
- Life Sciences, TÜBİTAK Marmara Research Center, Gebze-Kocaeli, Türkiye
| | | | - Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, Messina, Italy
| | - Abul Hossain
- Department of Biochemistry, Memorial University of Newfoundland, St. John 's, NL, Canada
| | - Fereidoon Shahidi
- Department of Biochemistry, Memorial University of Newfoundland, St. John 's, NL, Canada
| |
Collapse
|
786
|
Hu MX, Li JQ, Zhang HW, Ma YK, Zhao BJ, Xiao L, Liang W, Wang Y, Wang C, Wu CG, Liu TT. 1-benzyl-6-nitro-4-phenyl-4-(methoxycarbonyl)-2(1H)-pyridinone, a novel pirfenidone derivative, alleviate hepatic fibrosis through T cells. Biomed Pharmacother 2025; 184:117907. [PMID: 39978030 DOI: 10.1016/j.biopha.2025.117907] [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: 12/10/2024] [Revised: 01/17/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
Hepatic fibrosis (HF) is a pathological process in many liver diseases, which lack of specific agents. Pirfenidone (PFD) derivatives are potential new drug. The purpose of this study was to investigate the effect and immunological mechanism of PFD derivatives on HF. A total of 11 PFD derivatives were designed, synthesized and screened. 1-benzyl-6-nitro-4-phenyl-4-(methoxycarbonyl)-2(1 H)-pyridinone (code: Compound 5) had optimal effect on inhibiting nitric oxide release, hepatic stellate cells (HSCs) and T cell proliferation, which suggested that Compound 5 showed anti-inflammatory, anti-fibrosis and immunoregulation effects. Compound 5 inhibited the proliferation of HSC-T6 and T cell in dose-dependent manner, the IC50 was 10.19 μM and 17.16 μM, respectively. Compound 5 inhibited the differentiation of CD8+T cells and promoted the differentiation of Tregs in the splenic T lymphocyte of CCl4-induced mouse HF model. Besides, Compound 5 promoted HSC-T6 apoptosis in dose-dependent manner, accompanied by the down-regulation of α-smooth muscle actin (α-SMA) and collagen-I (Col-I). In terms of mechanism, Compound 5 had no significant effect on glucose uptake of T cells. But it inhibited non-esterified fatty acid (NEFA) secretion of T cell by inhibiting the phosphorylation of PI3K-AKT-mTOR signal, which related to the metabolism of T cell. Subsequently, Compound 5 affected α-SMA and Col-I expression of HSC-T6 by T cell modulating in cell co-culture. CONCLUSION: Compound 5 is a promising new drug against HF by the dual role of inhibiting HSCs and modulating T cells lipid metabolism, which affects the immune microenvironment of HF.
Collapse
Affiliation(s)
- Meng-Xue Hu
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, China; The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, China
| | - Jia-Qi Li
- School of Pharmacy, South-Central Minzu University, Wuhan, China
| | - Hong-Wei Zhang
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, China; The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, China
| | - Yu-Kun Ma
- Department of Pharmacy, the 902nd Hospital of the PLA Joint Logistics Support Force, Bengbu, China
| | - Bao-Jing Zhao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lei Xiao
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, China; The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, China
| | - Wei Liang
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, China; The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, China
| | - Yamin Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Chun Wang
- Institute of Clinical Pharmacology, Anhui Medical University, the Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Cheng-Gui Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China.
| | - Ting-Ting Liu
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, China; The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, China.
| |
Collapse
|
787
|
Shaheen MA, Darwish KM, Kishk SM, El-Sayed MAA, Salama I. Development of 1,2,3-triazole hybrids as multi-faced anticancer agents co-targeting EGFR/mTOR pathway and tubulin depolymerization. Bioorg Chem 2025; 156:108153. [PMID: 39855112 DOI: 10.1016/j.bioorg.2025.108153] [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: 11/12/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025]
Abstract
Novel 1,2,3-triazole hybrids bearing various substituents have been synthesized as potential anticancer agents. Ligand-based approach has been adopted to design these compounds relying on the hybridization of 1,2,3-triazole with α,β-unsaturated carbonyl, 5- and 6-membered heterocyclic scaffolds. All synthesized members were investigated for their cytotoxic potency against nine types comprising 60 panels of human cancerous cells by the US National Cancer Institute: Development Therapeutic Program (US_NCI_DTP). Among the tested members, 4b, 4e, and 4h showed prominent cytotoxic effects (> 80 % growth inhibition: GI) on a wide panel of tested cancer cell lines, mainly melanoma and colorectal cancer redeeming their selection for five dose testing. Presenting low nanomolar GI50 concentrations, two representative potent anticancer compounds 4b and 4e were subjected to cytotoxicity testing on colon normal cell (FHC) to investigate their safety window and they showed less toxicity to normal cells at the concentration required to produce anticancer effect. Furthermore, 4b and 4e were exposed to additional mechanistic studies in colorectal cancer cell HCT-116 suggesting multifaceted mechanisms of action. A study into the effects of cytotoxic chemicals 4b and 4e on cell cycle progression regulation showed triggered the arrest of cell cycles during the G1 and S phases. Moreover, 4b and 4e caused cell death mainly through apoptosis the thing that has been reinforced by the elevated Bax: Bcl2 ratio, as well as concentrations of caspases 3 and 9 within HCT-116. Further, both compounds showed prominent inhibition profiles against tubulin polymerization as well as EGFR catalytic activity reaching down to low-digit micromolar and sub-micromolar concentrations, respectively, as compared to positive reference controls. Compounds' impacts on gene expression of cancer-associated and EGFR-downstream signaling markers including TNFα, IL-6, and mTOR, were explored in HCT-116 highlighted significant downregulations versus the untreated cells. Docking studies demonstrated the specific fit of 4b and 4e into EGFR and the colchicine binding site of tubulin.
Collapse
Affiliation(s)
- Mennatallah A Shaheen
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522 Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University, Egypt.
| | - Khaled M Darwish
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522 Egypt; Medicinal Chemistry Department, Faculty of Pharmacy, Galala University, New Galala 43511 Egypt.
| | - Safaa M Kishk
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522 Egypt.
| | - Magda A-A El-Sayed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University, Egypt; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Ismail Salama
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522 Egypt.
| |
Collapse
|
788
|
Abulkhair HS, El-Adl K. A decade of research effort in synthesis, biological activity assessments, and mechanistic investigations of sulfamethazine-incorporating molecules. Arch Pharm (Weinheim) 2025; 358:e2500033. [PMID: 40123425 DOI: 10.1002/ardp.202500033] [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: 01/12/2025] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
Because of its importance in medicinal chemistry, scientific researchers have been interested in incorporating sulfamethazine in developing biologically active candidates. To achieve this, several synthetic approaches have been adopted. The adopted approaches included condensation with electrophilic reactants, coupling with nucleophilic aromatics and active methylene compounds, Knoevenagel condensation, Doebner Miller reaction, microwave-assisted click cycloaddition, green reaction routes, and multicomponent reaction. Linking this molecular scaffold to a variety of heterocycles in the last 10 years furnished a set of potential anti-inflammatory, antiviral, anticancer, antiparkinsonian, neuroprotective, and antidiabetic candidates targeting H5N1 NA, epidermal growth factor receptor, acetylcholinesterase (AChE), butylcholinesterase (BChE), human carbonic anhydrase (hCA), α-amylase, and α-glucosidase. This review reports all the adopted synthetic approaches, the biological activities studied, structure-activity relationship analyses, and the mechanistic investigations of the reported organic sulfamethazine-incorporating molecules throughout 2015-2024, based on information retrieved from three search engines: Scopus, PubMed, and Google Scholar.
Collapse
Affiliation(s)
- Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| | - Khaled El-Adl
- Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, Egypt
| |
Collapse
|
789
|
Benedik NS, Proj M, Steinebach C, Sova M, Sosič I. Targeting TAK1: Evolution of inhibitors, challenges, and future directions. Pharmacol Ther 2025; 267:108810. [PMID: 39909209 DOI: 10.1016/j.pharmthera.2025.108810] [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: 11/15/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
The increasing incidence of inflammatory and malignant diseases signifies the need to develop first-in-class drugs with novel mechanisms of action. In this respect, the transforming growth factor (TGF)-β-activated kinase 1 (TAK1), an essential part of several signaling pathways, is considered relevant and promising. This manuscript provides a brief overview of the signal transduction orchestrated by TAK1 within these pathways, followed by an in-depth and thorough analysis of the chemical matter demonstrated to inhibit this kinase. Special attention is given to the selectivity profiling of inhibitors, as well as to the outcomes of their biological characterization. Because published TAK1 inhibitors differ significantly in their kinome selectivity, active-site binding, and biological activity, we hope that this review will allow a judicial estimation of their quality and usefulness for TAK1-addressing assays. Our thoughts on the perspectives and possible developments of the field are also provided to assist scientists who are involved in the design and development of TAK1-targeting modulators.
Collapse
Affiliation(s)
- Nika Strašek Benedik
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Matic Proj
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Christian Steinebach
- Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Matej Sova
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Izidor Sosič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
790
|
Le MHN, Nguyen PK, Nguyen TPT, Nguyen HQ, Tam DNH, Huynh HH, Huynh PK, Le NQK. An in-depth review of AI-powered advancements in cancer drug discovery. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167680. [PMID: 39837431 DOI: 10.1016/j.bbadis.2025.167680] [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: 10/18/2024] [Revised: 01/12/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
The convergence of artificial intelligence (AI) and genomics is redefining cancer drug discovery by facilitating the development of personalized and effective therapies. This review examines the transformative role of AI technologies, including deep learning and advanced data analytics, in accelerating key stages of the drug discovery process: target identification, drug design, clinical trial optimization, and drug response prediction. Cutting-edge tools such as DrugnomeAI and PandaOmics have made substantial contributions to therapeutic target identification, while AI's predictive capabilities are driving personalized treatment strategies. Additionally, advancements like AlphaFold highlight AI's capacity to address intricate challenges in drug development. However, the field faces significant challenges, including the management of large-scale genomic datasets and ethical concerns surrounding AI deployment in healthcare. This review underscores the promise of data-centric AI approaches and emphasizes the necessity of continued innovation and interdisciplinary collaboration. Together, AI and genomics are charting a path toward more precise, efficient, and transformative cancer therapeutics.
Collapse
Affiliation(s)
- Minh Huu Nhat Le
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan
| | - Phat Ky Nguyen
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan.
| | | | - Hien Quang Nguyen
- Cardiovascular Research Department, Methodist Hospital, Merrillville, IN 46410, USA
| | - Dao Ngoc Hien Tam
- Regulatory Affairs Department, Asia Shine Trading & Service Co. LTD, Viet Nam
| | - Han Hong Huynh
- International Master Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Phat Kim Huynh
- Department of Industrial and Systems Engineering, North Carolina A&T State University, Greensboro, NC 27411, USA.
| | - Nguyen Quoc Khanh Le
- AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan; In-Service Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan.
| |
Collapse
|
791
|
Bhatia N, Kumar S, Goyal LD, Thareja S. Optimizing selective estrogen receptor degraders for anticancer drug development. Future Med Chem 2025; 17:637-640. [PMID: 40008677 PMCID: PMC11938951 DOI: 10.1080/17568919.2025.2467615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Affiliation(s)
- Neha Bhatia
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences Central University of Punjab, Bathinda, India
| | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, India
| | - Lajya Devi Goyal
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, Bathinda, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences Central University of Punjab, Bathinda, India
| |
Collapse
|
792
|
Mohan Malothu R, Thalari G. Design and Synthesis of Novel Thiazole Linked Tetrahydropyridine Analogues as Anticancer Agents. Chem Biodivers 2025; 22:e202402223. [PMID: 39523463 DOI: 10.1002/cbdv.202402223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
A library of new thiazole linked tetrahydropyridines (6a-q) synthesized and screened for their invitro anticancer activity against human breast adeno carcinoma cell viz. MCF-7 and MDA-MB-231. The two compounds 6d containing -F and -Cl functions in para and meta position of phenyl ring (9.94±1.02 μM, 9.78±1.08 μM) and 6e with -Cl and -NH2 functions on pyridine ring (9.72±0.91 μM, 9.54±0.95 μM) demonstrated outstanding activity against both the cell lines when compared to Doxorubicin. The benzofuran analogue 6o presented good activity with an IC50 value of 12.19±1.03 μM (MCF-7) and 12.22±1.07 μM (MDA-MB-231). The molecular docking study of potent molecule 6e against crystal structure of breast tumor kinase presented promising docking score and binding interactions. Predicted pharmacokinetics properties of compounds 6a-q and presented boiled diagram of compounds 6d and 6e implied favourable drug-likeness properties.
Collapse
Affiliation(s)
- Ram Mohan Malothu
- Natural Products Laboratory, Department of Chemistry, University College of Science, Osmania University, 500007, Hyderabad, Telangana, India
- Aragen Life Sciences, Nacharam, Hyderabad, Telangana, India
| | - Gangadhar Thalari
- Natural Products Laboratory, Department of Chemistry, University College of Science, Osmania University, 500007, Hyderabad, Telangana, India
| |
Collapse
|
793
|
Cao M, Nguyen T, Song J, Zheng YG. Biomedical effects of protein arginine methyltransferase inhibitors. J Biol Chem 2025; 301:108201. [PMID: 39826691 PMCID: PMC11871472 DOI: 10.1016/j.jbc.2025.108201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/22/2025] Open
Abstract
Protein arginine methyltransferases (PRMTs) are enzymes that catalyze the methylation of arginine residues in eukaryotic proteins, playing critical roles in modulating diverse cellular processes. The importance of PRMTs in the incidence and progression of a wide range of diseases, particularly cancers, such as breast, liver, lung, colorectal cancer, lymphoma, leukemia, and acute myeloid leukemia is increasingly recognized. This underscores the critical need for the development of effective PRMT inhibitors as therapeutic intervention. The field of PRMT inhibitors is in the rapidly growing phase and it is necessary to conduct a summative review of how the so-far developed inhibitors impact PRMT functions and cellular physiology. Our review aims to summarize molecular action mechanisms of these PRMT inhibitors and particularly elaborate their triggered biomedical effects. We describe the cellular phenotype consequences of select PRMT inhibitors across various disease models, thereby providing an understanding of the pharmacological mechanisms underpinning PRMT inhibition. The promising effects of PRMT5 inhibitors in targeted therapy of methylthioadenosine phosphorylase-deleted cancers are particularly highlighted. At last, we provide a perspective on the challenges and further opportunities of developing and applying novel PRMT inhibitors for clinical advancement.
Collapse
Affiliation(s)
- Mengtong Cao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia, United States
| | - Terry Nguyen
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia, United States
| | - Jiabao Song
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia, United States
| | - Y George Zheng
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia, United States.
| |
Collapse
|
794
|
Schwartz J, Capistrano K, Hussein H, Hafedi A, Shukla D, Naqvi A. Oral SARS-CoV-2 Infection and Risk for Long Covid. Rev Med Virol 2025; 35:e70029. [PMID: 40074704 PMCID: PMC11903386 DOI: 10.1002/rmv.70029] [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: 09/18/2024] [Revised: 11/30/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025]
Abstract
SARS-CoV-2 is an oral pathogen that infects and replicates in mucosal and salivary epithelial cells, contributing to oral post-acute sequelae COVID-19 (PASC) and other oral and non-oral pathologies. While pre-existing inflammatory oral diseases provides a conducive environment for the virus, acute infection and persistence of SARS-CoV-2 can also results in oral microbiome dysbiosis that further worsens poor oral mucosal health. Indeed, oral PASC includes periodontal diseases, dysgeusia, xerostomia, pharyngitis, oral keratoses, and pulpitis suggesting significant bacterial contributions to SARS-CoV-2 and oral tissue tropism. Dysbiotic microbiome-induced inflammation can promote viral entry via angiotensin-converting enzyme receptor-2 (ACE2), serine transmembrane TMPRSS2 and possibly other non-canonical pathways. Additionally, metabolites derived from a dysbiotic microbiome can alter the physiological and biochemical pathways related to the metabolism of lipids, carbohydrates, and amino acids. This may promote a pro-inflammatory microenvironment, leading to immune exhaustion, loss of tolerance, and susceptibility to a variety of oral pathogens, causing acute and later chronic inflammation. Microbial release of mimics of host metallopeptidases related to furin, ADAM17 (A disintegrin and metalloproteinase 17), and glycoprotein metabolites can further aid viral attachment to T cell immunoglobulin-like (TIMs), enhancing viral entry while simultaneously depressing oral mucosal immune resistance and clearance. Membrane reorganization characterised by neuroproteins, such as neuropilins, also functionally assists with SARS-CoV-2 entry and extends the pathogenesis of PASC from the oral cavity to the brain, gut, or other non-oral tissues. Thus, poor oral health, characterised by disrupted oral microbiomes can promote viral tropism, weaken antiviral resistance, and heightens susceptibility to SARS-CoV-2 infection. This immune dysfunction also increases the risk of additional viral infections, exacerbating oral conditions like periodontal and endodontic diseases. These persistent oral health issues can contribute to systemic inflammation, creating bidirectional effects between oral and non-oral tissues, potentially leading to Post-Acute Sequelae of COVID-19 (PASC).
Collapse
Affiliation(s)
- Joel Schwartz
- Department of Oral Medicine and Diagnostic SciencesUniversity of Illinois ChicagoChicagoIllinoisUSA
| | | | - Heba Hussein
- Department of Oral MedicineFaculty of DentistryCairo UniversityCairoEgypt
| | - Avin Hafedi
- Department of PeriodonticsUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - Deepak Shukla
- Department of Microbiology and ImmunologyUniversity of Illinois ChicagoChicagoIllinoisUSA
- Department of Ophthalmology and Visual SciencesUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - Afsar Naqvi
- Department of PeriodonticsUniversity of Illinois ChicagoChicagoIllinoisUSA
- Department of Microbiology and ImmunologyUniversity of Illinois ChicagoChicagoIllinoisUSA
| |
Collapse
|
795
|
Lee Y, Ahn S, Jung E, Lim Y, Koh D, Bae DH, Shin SY. (E)-3-(3-([1,1'-Biphenyl]-4-yl)-1-phenyl-1H-pyrazol-4-yl)-1-phenylprop-2-en-1-ones inducing reactive oxygen species generation through glutathione depletion. Bioorg Med Chem Lett 2025; 117:130043. [PMID: 39608685 DOI: 10.1016/j.bmcl.2024.130043] [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: 09/05/2024] [Revised: 11/13/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
The accumulation of reactive oxygen species (ROS) disrupts reduction-oxidation homeostasis, which can result in damage to cancer cells. To identify the compounds generating ROS, compounds containing Michael acceptors were designed because they are suggested to be critical for ROS elevation via glutathione depletion. Twelve (E)-3-(3-([1,1'-biphenyl]-4-yl)-1-phenyl-1H-pyrazol-4-yl)-1-phenylprop-2-en-1-ones were synthesized and identified using nuclear magnetic resonance spectroscopy and mass spectrometry. Intracellular ROS levels induced by treatment with the compounds were determined using fluorescence microscopy with the oxidant-sensing fluorescent probe 2',7'-dichlorodihydrofluorescein diacetate. We selected compound 9, which showed the highest activity, and performed further biological experiments, including glutathione depletion and apoptosis assays, using MIA PaCa-2 pancreatic cancer cells. Additionally, the reason why the intracellular ROS level by compound 9 was lower than that of menadione used as a control was explained through in silico docking experiments. Our findings suggest that compound 9 has the potential to act as an anticancer agent by inducing ROS generation through the depletion of intracellular glutathione.
Collapse
Affiliation(s)
- Youngshim Lee
- Division of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Seunghyun Ahn
- Department of Applied Chemistry, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Euitaek Jung
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Yoongho Lim
- Division of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Dongsoo Koh
- Department of Applied Chemistry, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Dong-Ho Bae
- Department of Food Sciences and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
796
|
Al-Karmalawy AA, Zeidan MA, Elmaaty AA, Sharaky M, Yassen ASA, Khaleel EF, Eldehna WM, Ashour HF. Design and synthesis of new 1,2,3-triazole derivatives as VEGFR-2/telomerase downregulatory candidates endowed with apoptotic potential for cancer treatment. Bioorg Chem 2025; 156:108159. [PMID: 39823817 DOI: 10.1016/j.bioorg.2025.108159] [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: 11/06/2024] [Revised: 12/15/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025]
Abstract
In this current work, we dedicated efforts to designing and synthesizing new 1,2,3-triazole-analogues (5a-d), (6a-d), and (7a-c) to act as dual VEGFR-2 and telomerase inhibitors with promising apoptotic potential. The synthesized analogues were examined against eleven diverse types of cancer cells and two normal cells to assess their ability to inhibit cell growth (GI%). Obviously, compound 7b showed the best average GI% (75.69 %) surpassing the average GI% of Dox (65.79 %). Compound 5d showed the lowest IC50 values (25.86 and 51.91 µM) against HNO-97 and FaDu cancer cells, respectively. Besides, compound 5a exhibited the lowest IC50 value (15.46 µM) against HCT116, whereas compound 6b revealed the lowest IC50 value (31.14 µM) against HuH7. Besides, candidates 5a, 5b, 5d, and 7a showed prominent inhibitory results towards VEGFR-2 protein with decreasing its expression by 0.33, 0.42, 0.38 and 0.26-fold change, respectively. However, compounds 5a, 5b, 5d, and 7a showed promising inhibitory results towards telomerase protein and decreased its expression by 0.60, 0.50, 0.52, and 0.44-fold change, respectively. Additionally, it was clear that compound 5a was able to upregulate the expression of Caspases 3, 8, and 9 proteins by 2.19, 1.83, and 1.62-fold change, respectively. Besides, 5a was able to downregulate the expression of CDK-2, CDK-4, and CDK-6 proteins by 0.50, 0.43, and 0.13-fold change, respectively. Obviously, compound 5a halted the cell cycle at the G1, S, and G2-M phases in HCT116 cells. Subsequently, the synthesized 1,2,3-triazole analogues can be treated as lead VEGFR-2 and telomerase inhibitors with potential apoptotic activity for future optimization and cancer treatment.
Collapse
Affiliation(s)
- Ahmed A Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad 10023, Iraq; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt.
| | - Mohamed A Zeidan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Ayman Abo Elmaaty
- Medicinal Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt; Medicinal Chemistry Department, Clinical Pharmacy Program, East Port Said National University, Port Said 42526, Egypt
| | - Marwa Sharaky
- Cancer Biology Department, Pharmacology Unit, National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | - Asmaa S A Yassen
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt
| | - Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Asir 61421, Saudi Arabia
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648, Egypt
| | - Heba F Ashour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt
| |
Collapse
|
797
|
Subramani T, Saravanan H, David H, Solanke J, Rajaramon S, Dandela R, Solomon AP. Bioorganic compounds in quorum sensing disruption: strategies, Mechanisms, and future prospects. Bioorg Chem 2025; 156:108192. [PMID: 39874908 DOI: 10.1016/j.bioorg.2025.108192] [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: 08/16/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
Recent research has shed light on the complex world of bacterial communication through quorum sensing. This sophisticated intercellular signalling mechanism, driven by auto-inducers, regulates crucial bacterial community behaviours such as biofilm formation, expression of virulence factors, and resistance mechanisms. The increasing threat of antibiotic resistance, coupled with quorum sensing mediated response, necessitates alternative strategies to combat bacterial infections. Quorum quenching has emerged as a promising approach, utilizing quorum quenching enzymes and quorum sensing inhibitors to disrupt quorum sensing signalling pathways, thus reducing virulence and biofilm formation. This review focuses on natural and synthetic bioorganic compounds that act as quorum-sensing inhibitors, providing insights into their mechanisms, structure-activity relationships, and potential as anti-virulence agents. The review also explores the communication languages of bacteria, including AHLs in gram-negative bacteria, oligopeptides in gram-positive bacteria, and LuxS, a universal microbial language. By highlighting recent advancements and prospects in bioorganic QSIs, this article underscores their crucial role in developing effective anti-virulence therapies and combating the growing threat of antimicrobial resistance.
Collapse
Affiliation(s)
- Tarunkarthick Subramani
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Harish Saravanan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Jayshree Solanke
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Bhubaneswar, Odisha, India
| | - Shobana Rajaramon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India.
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Bhubaneswar, Odisha, India.
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India.
| |
Collapse
|
798
|
Haider MK, Ullah A, Gopiraman M, Kim IS. A review on bioactivity, plant safety, and metal-reducing potential of lignin, its micro/nanostructures, and composites. Int J Biol Macromol 2025; 294:139366. [PMID: 39743055 DOI: 10.1016/j.ijbiomac.2024.139366] [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: 10/29/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Modern science focuses on sustainability-oriented innovation. Structurally sophisticated lignin is a sustainable alternative to non-renewable resources. Over the last several years, a tremendous scientific effort has been made to innovate lignin-based sustainable materials for numerous advanced applications. The lignin's phenolic, methoxyl and aliphatic hydroxyl functional groups are biologically and chemically active, making it conducive to developing state-of-the-art biomedicine, food packaging, crop protection, and catalyst materials. The biocidal effect of lignin rests on the phenolic compounds, specifically the double bond in α, β positions of the side chain, and a methyl group in the γ position. Also, depending on the biomass source and the pulping method, lignins possess different biocidal and antioxidant properties. The abundant hydroxyl groups in lignin are metal reductants and possess capping ability for the nanoparticles (NPs). This review focused on lignin's bioactivity mechanism, including antimicrobial efficacy and antioxidant properties. Lignin-based micro/nanocomposites and their application on food packaging, plant protection, and growth will also be explored. We will also review the application of lignin as a reducing and capping agent for the synthesis of metal NPs.
Collapse
Affiliation(s)
- Md Kaiser Haider
- Nano Fusion Technology Research Group, Institute for Fiber Engineering and Science (IFES), Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Tokida 3-15-1, Ueda, Nagano 386-8567, Japan
| | - Azeem Ullah
- Nano Fusion Technology Research Group, Institute for Fiber Engineering and Science (IFES), Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Tokida 3-15-1, Ueda, Nagano 386-8567, Japan
| | - Mayakrishnan Gopiraman
- Nano Fusion Technology Research Group, Institute for Fiber Engineering and Science (IFES), Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Tokida 3-15-1, Ueda, Nagano 386-8567, Japan
| | - Ick Soo Kim
- Nano Fusion Technology Research Group, Institute for Fiber Engineering and Science (IFES), Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Tokida 3-15-1, Ueda, Nagano 386-8567, Japan.
| |
Collapse
|
799
|
Hellmann A, Zwara A, Weryszko O, Czapiewska M, Korczynska J, Sztendel A, Śledziński T, Mika A. Evaluation of the effect of Hashimoto's Thyroiditis on fatty acids involved in inflammation in the thyroid tissue. Biomed Pharmacother 2025; 184:117894. [PMID: 39913970 DOI: 10.1016/j.biopha.2025.117894] [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: 10/31/2024] [Revised: 01/16/2025] [Accepted: 02/03/2025] [Indexed: 03/04/2025] Open
Abstract
Hashimoto's thyroiditis (HT) is one of the most common autoimmune diseases associated with inflammation of the thyroid gland. Some fatty acids (FAs) are involved in inflammation. The aim of the study was to investigate how HT affects FA metabolism in thyroid tissue. We examined normal thyroid tissue from 92 patients with and without HT. We determined total FA panel, pro-inflammatory and specialized pro-resolving lipid mediators (SPMs) concentrations and measured the expression levels of genes encoding the corresponding enzymes of FA metabolism and mediator's synthesis. In HT tissue, we observed increased total lipid levels (p < 0.05), decreased carnitine palmitoyltransferase I (p < 0.05) and increased tumor necrosis factor alpha (p < 0.001) compared to healthy tissue. Simultaneously, we observed an overexpression of enzymes responsible for the synthesis of polyunsaturated FAs and their higher levels in HT tissue. However, despite the overexpression of phospholipase A2 (p < 0.001), FA translocase CD36 (p < 0.05) and higher levels of free arachidonic acid in HT tissue (p < 0.05), we observed no differences in the expression of COX-2 in both tissues and, interestingly, a downregulation of 15-LOX (p = 0.001) and lower concentrations of SPMs. Finally, the opposite effect of HT on enzymes responsible for anti-inflammatory BCFAs synthesis in mitochondria and cytosol could indicate a compensatory mechanism. To summarize, the mechanism of the effect of HT on FA metabolism is certainly complex. Thyrocytes are perturbed by the inflammatory effects associated with HT, leading to mitochondrial dysfunction and consequently decreased β-oxidation and BCFA metabolism in mitochondria. Furthermore, despite a significant synthesis of PUFAs, SPMs are also not produced.
Collapse
Affiliation(s)
- Andrzej Hellmann
- Department of Department of Surgical Oncology, Transplant Surgery and General Surgery, Faculty of Medicine, Medical University of Gdansk, Smoluchowskiego 17, Gdansk 80-214, Poland
| | - Agata Zwara
- Department of Environmental Analytics, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk 80-308, Poland
| | - Oliwia Weryszko
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Monika Czapiewska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Justyna Korczynska
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Alicja Sztendel
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Tomasz Śledziński
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland
| | - Adriana Mika
- Department of Environmental Analytics, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk 80-308, Poland; Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki, Gdansk 80-211, Poland.
| |
Collapse
|
800
|
Sharma V, Kumar A. MCL-1 as a potent target for cancer: Recent advancements, structural insights and SAR studies. Bioorg Chem 2025; 156:108211. [PMID: 39889551 DOI: 10.1016/j.bioorg.2025.108211] [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: 12/07/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
The myeloid cell leukemia-1 (Mcl-1) differentiation protein belongs to the B-cell lymphoma 2 (Bcl-2) family of proteins which regulates the apoptosis or cell death. Mcl-1 is known for its pro-survival in response to various stressors. Therefore, it acts as a prominent target in cancer treatment. Mcl-1 has emerged as one of the validated drug targets for anticancer drug discovery as their expression has been implicated in the pathogenesis of cancers. In this review, we have included the various inhibitors based on many heterocyclic rings such as pyrrole, pyrazole, coumarin, quinoline and indole. This manuscript incorporates the anticancer activity, structure activity relationship (SAR) and molecular modelling of recently synthesized Mcl-1 inhibitors. The clinical trial status of Mcl-1 inhibitors is also described. But till now, no Mcl-1 inhibitor has been approved by any drug authority. This review is based on extensive research in the field of designing Mcl-1 inhibitors from 2020 to till now. It will provide extensive information to researchers and scientists for designing of novel Mcl-1 inhibitors.
Collapse
Affiliation(s)
- Vishakha Sharma
- Amity School of Pharmaceutical Sciences, Amity University Punjab, Mohali, India
| | - Ankush Kumar
- Amity School of Pharmaceutical Sciences, Amity University Punjab, Mohali, India.
| |
Collapse
|