51
|
Duan Y, Liu Z, Wang Q, Zhang J, Liu J, Zhang Z, Li C. Targeting MYC: Multidimensional regulation and therapeutic strategies in oncology. Genes Dis 2025; 12:101435. [PMID: 40290126 PMCID: PMC12022651 DOI: 10.1016/j.gendis.2024.101435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/05/2024] [Accepted: 08/25/2024] [Indexed: 04/30/2025] Open
Abstract
MYC is dysregulated in approximately 70% of human cancers, strongly suggesting its essential function in cancer. MYC regulates many biological processes, such as cell cycle, metabolism, cellular senescence, apoptosis, angiogenesis, and immune escape. MYC plays a central role in carcinogenesis and is a key regulator of tumor development and drug resistance. Therefore, MYC is one of the most alluring therapeutic targets for developing cancer drugs. Although the search for direct inhibitors of MYC is challenging, MYC cannot simply be assumed to be undruggable. Targeting the MYC-MAX complex has been an effective method for directly targeting MYC. Alternatively, indirect targeting of MYC represents a more pragmatic therapeutic approach, mainly including inhibition of the transcriptional or translational processes of MYC, destabilization of the MYC protein, and blocking genes that are synthetically lethal with MYC overexpression. In this review, we delineate the multifaceted roles of MYC in cancer progression, highlighting a spectrum of therapeutic strategies and inhibitors for cancer therapy that target MYC, either directly or indirectly.
Collapse
Affiliation(s)
- Yingying Duan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Zhaoshuo Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Qilin Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Junyou Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Jiaxin Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Ziyi Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Chunyan Li
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Key Laboratory of Big Data-Based Precision Medicine (Ministry of Industry and Information Technology), Beihang University, Beijing 100191, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100191, China
| |
Collapse
|
52
|
Ur Rahman M, Hussain HR, Akram H, Sarfraz M, Nouman M, Khan JA, Ishtiaq M. Niosomes as a targeted drug delivery system in the treatment of breast cancer: preparation, classification and mechanisms of cellular uptake. J Drug Target 2025; 33:916-932. [PMID: 39964023 DOI: 10.1080/1061186x.2025.2468750] [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: 09/27/2024] [Revised: 01/24/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
Breast cancer (BC) remains one of the significant health issues across the globe, being diagnosed in millions of women worldwide annually. Conventional therapeutic options have substantial adverse effects due to their non-specificity and limited drug bioavailability. Niosomes, being novel drug delivery systems formed from non-ionic surfactants, with or without cholesterol and charge-inducing agents, are used as therapeutic options in treating BC. Their formulation by various methods enhances the therapeutic efficacy and bioavailability and minimises side effects. Niosomal formulation of tamoxifen exhibits target drug delivery with enhanced stability, whereas docetaxel and methotrexate show sustained and controlled drug release, respectively. 5-Fluorouracil, doxorubicin, paclitaxel, cyclophosphamide and epirubicin show improved cytotoxic effects against BC when combined with other agents. Furthermore, repurposed niosomal formulations of anti-cancer drugs show improved penetration, reduced tumour volume and significantly enhanced anti-tumour effect. This review article focuses on the composition of niosomes and their application in BC treatment and then examines how niosomes could contribute to BC research.
Collapse
Affiliation(s)
| | | | - Habiba Akram
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Sarfraz
- College of Pharmacy, Al-Ain University, Al-Ain, United Arab Emirates
| | - Muhammad Nouman
- College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Jawad Akbar Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore, Pakistan
| | - Memona Ishtiaq
- Department of Pharmacy, Lahore Institute of Professional Studies, Lahore, Pakistan
| |
Collapse
|
53
|
Alsaidan OA. Recent advancements in aptamers as promising nanotool for therapeutic and diagnostic applications. Anal Biochem 2025; 702:115844. [PMID: 40090606 DOI: 10.1016/j.ab.2025.115844] [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: 12/17/2024] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Aptamers are single-strand oligonucleotide molecules having certain structural interactions which allow them to bind to specific targets. Modified nucleotides are added during or after a selection procedure like Systematic Evolution of Ligands by Exponential Enrichment i.e., SELEX to enhance the characteristics and functionality of aptamers. Aptamers are extensible molecular tools with several uses such as in drug administration, biosensing, bioimaging, drug therapies and diagnostics. The ability to detect is improved by using aptamer-based sensors in conjunction with biological molecules among other sensing techniques. Chemical modification, and strong resistance to denaturation, aptamers are appropriate biological recognizing agents for developing sensitive and repeatable aptasensors. This review discusses the most current developments in the aptamers, SELEX method, applications of aptamers as innovative diagnostic, therapeutic & theragnostic tool along with major limitations & prospective directions in the future.
Collapse
Affiliation(s)
- Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, P.O. Box 2014, Sakaka, 72341, Saudi Arabia.
| |
Collapse
|
54
|
Nörnberg AB, Gularte MS, da Fonseca CAR, San José DEJM, Wilhelm EA, Fajardo AR. 3D-printed gelatin/dialdehyde starch hydrogels for hydrocortisone topical administration and in vivo treatment of atopic dermatitis. Colloids Surf B Biointerfaces 2025; 251:114613. [PMID: 40073627 DOI: 10.1016/j.colsurfb.2025.114613] [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: 02/15/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Atopic dermatitis (AD) is a chronic disorder affecting millions worldwide. Recent advancements suggest that combining therapies can significantly improve AD treatment outcomes and mitigate the challenges of long-term drug use, particularly with corticosteroids. In this study, we developed a 3D-printed hydrogel composed of gelatin (Gel) and dialdehyde starch (DAS), capable of encapsulating and delivering hydrocortisone (HC). DAS was synthesized via an oxidation reaction, introducing aldehyde groups that facilitated hydrogel formation with Gel through imine bond formation (Schiff base reaction). By adjusting the Gel/DAS ratio, we formulated inks with suitable rheological properties for extrusion-based 3D printing, an approach not yet fully exploited in this context. The resulting Gel/DAS network successfully encapsulated HC, as demonstrated by characterization analyses. The printed hydrogel exhibited a well-defined microstructure, significant water absorption and retention capabilities, and excellent stability. HC release followed a controlled mechanism consistent with Korsmeyer-Peppas kinetics. In an in vivo model, the 3D-printed hydrogel containing HC showed therapeutic efficacy comparable to conventional HC treatments in alleviating AD symptoms in mice. Additionally, the hydrogel significantly reduced myeloperoxidase (MPO) activity and increased non-protein thiol (NPSH) levels in the dorsal skin of DNCB-exposed mice, underscoring its therapeutic potential.
Collapse
Affiliation(s)
- Andressa B Nörnberg
- Laboratório de Tecnologia e Desenvolvimento de Compósitos e Materiais Poliméricos (LaCoPol), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão s/n, Pelotas, RS 96010-900, Brazil
| | - Matheus S Gularte
- Laboratório de Tecnologia e Desenvolvimento de Compósitos e Materiais Poliméricos (LaCoPol), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão s/n, Pelotas, RS 96010-900, Brazil
| | - Caren A R da Fonseca
- Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS 96010-900, Brazil
| | - Daniel E J M San José
- Laboratório de Tecnologia e Desenvolvimento de Compósitos e Materiais Poliméricos (LaCoPol), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão s/n, Pelotas, RS 96010-900, Brazil
| | - Ethel A Wilhelm
- Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS 96010-900, Brazil
| | - André R Fajardo
- Laboratório de Tecnologia e Desenvolvimento de Compósitos e Materiais Poliméricos (LaCoPol), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão s/n, Pelotas, RS 96010-900, Brazil.
| |
Collapse
|
55
|
He J, Illingworth H, Ullrich S, Ghosh P, Ton J, Jackson CJ, Nitsche C. Synthesis, screening and validation of cysteine-reactive fragments as chikungunya virus protease inhibitors. Bioorg Med Chem Lett 2025; 122:130176. [PMID: 40081602 DOI: 10.1016/j.bmcl.2025.130176] [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: 01/09/2025] [Revised: 02/21/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Alphaviruses like the Chikungunya virus cause severe outbreaks; however, no specific treatments are available. Their viral replication depends on the nsP2 cysteine protease, a promising but underexplored target for drug discovery. In this study, we report a covalent fragment screening against Chikungunya virus nsP2 protease, resulting in the identification of three inhibitors that can serve as starting points for future drug development. Careful validation proved indispensable in eliminating false-positive hits from a Förster resonance energy transfer (FRET)-based inhibition assay, wherein interference was caused by the inner filter effect between the fluorescent substrate and coloured compounds. Jump-dilution experiments accompanied by reactivity studies with cysteine and the recombinant protein indicate covalent inhibition via thia-Michael addition. We further demonstrate cross-inhibition of the related alphavirus nsP2 protease from Sindbis virus. The study provides early insights into nsP2 inhibition by electrophilic fragments featuring non-promiscuous N-arylacrylamides, thus advancing the search for antivirals targeting Chikungunya and other alphaviruses of concern.
Collapse
Affiliation(s)
- Junming He
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Henry Illingworth
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Sven Ullrich
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Pritha Ghosh
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Jennifer Ton
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Colin J Jackson
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia; Australian Research Council Centre of Excellence in Synthetic Biology, Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
| | - Christoph Nitsche
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|
56
|
Roy S, Pramanik P, Bhattacharya S. Exploring the role of G-quadruplex DNA, and their structural polymorphism, in targeting small molecules for the design of anticancer therapeutics: Progress, challenges, and future directions. Biochimie 2025; 234:120-145. [PMID: 40250703 DOI: 10.1016/j.biochi.2025.04.004] [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: 09/27/2024] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Selective stabilization of non-canonical G-quadruplex DNA structures by small molecules can be a potential target for anticancer therapeutics. The primary motivation for the molecular design of these G-quadruplex binders is to restrict the transcriptional machinery, which can impede cancer cell progression. This review article comprises the structural diversity of different G-quadruplex DNA, the design strategy for targeting these structures with small molecules, and various G-quadruplex binding ligands which have been expanded by the chemists and biologists over the past few decades. Further, the existence of G-quadruplex structures inside human cells, the significant challenges for designing these selective G-quadruplex binding ligands, current status, and progress towards achieving this goal have also been discussed.
Collapse
Affiliation(s)
- Soma Roy
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India; School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India
| | - Pulakesh Pramanik
- School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India
| | - Santanu Bhattacharya
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India; School of Applied & Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Kolkata, 700032, India; Technical Research Centre, Indian Association for the Cultivation of Science, Kolkata, 700032, India; Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati, 517619, India.
| |
Collapse
|
57
|
Liu A, Zhu XJ, Sun WD, Bi SZ, Zhang CY, Lai SY, Li JH. Nicotinamide N-methyltransferase as a potential therapeutic target for neurodegenerative disorders: Mechanisms, challenges, and future directions. Exp Neurol 2025; 389:115253. [PMID: 40221009 DOI: 10.1016/j.expneurol.2025.115253] [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: 01/31/2025] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
Neurodegenerative diseases (NDs), including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), are characterized by progressive neuronal loss and functional decline, posing significant global health challenges. Emerging evidence highlights nicotinamide N-methyltransferase (NNMT), a cytosolic enzyme regulating nicotinamide (NAM) methylation, as a pivotal player in NDs through its dual impact on epigenetic regulation and metabolic homeostasis. This review synthesizes current knowledge on NNMT's role in disease pathogenesis, focusing on its epigenetic modulation via DNA hypomethylation and histone modifications, alongside its disruption of NAD+ synthesis and homocysteine (Hcy) metabolism. Elevated NNMT activity depletes NAD+, exacerbating mitochondrial dysfunction and impairing energy metabolism, while increased Hcy levels drive oxidative stress, neuroinflammation, and aberrant protein aggregation (e.g., Aβ, tau, α-synuclein). Notably, NNMT overexpression in AD and PD correlates with neuronal hypomethylation and neurotoxicity, as observed in postmortem brain studies and transgenic models. Mechanistically, NNMT consumes S-adenosylmethionine (SAM), limiting methyl donor availability for DNA methyltransferases (DNMTs) and histone methyltransferases (HMTs), thereby altering gene expression patterns critical for neuronal survival. Concurrently, NNMT-mediated NAD+ depletion disrupts sirtuin activity (e.g., SIRT1) and mitochondrial biogenesis, accelerating axonal degeneration. Therapeutic strategies targeting NNMT, such as RNA interference (RNAi), small-molecule inhibitors and exercise therapy, show promise in preclinical models by restoring NAD+ levels and reducing Hcy toxicity. However, challenges persist in achieving cellular specificity, optimizing blood-brain barrier penetration, and mitigating off-target effects. This review underscores NNMT's potential as a multifactorial therapeutic target, bridging metabolic and epigenetic dysregulation in NDs. Future research should prioritize elucidating tissue-specific NNMT interactions, refining inhibitor pharmacokinetics, and validating translational efficacy in clinical trials. Addressing these gaps could pave the way for novel disease-modifying therapies to combat the rising burden of neurodegeneration.
Collapse
Affiliation(s)
- An Liu
- Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Xiao-Juan Zhu
- Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Wei-Dong Sun
- Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Shuang-Zhou Bi
- Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Chen-Ying Zhang
- Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Shi-Yan Lai
- Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China
| | - Jiang-Hua Li
- Physical Education College, Jiangxi Normal University, Nanchang 330022, Jiangxi Province, China.
| |
Collapse
|
58
|
Saeed M, Elsadek MA, Chen Z, Zhao L, Wang G, Zhou C, Sun D, Gao Z, Jiao Y. Enhancing the terpenoid and flavonoid profiles and fruit quality in an elite Chinese bayberry line through hybridization. Food Chem 2025; 479:143784. [PMID: 40081069 DOI: 10.1016/j.foodchem.2025.143784] [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: 11/11/2024] [Revised: 02/19/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Chinese bayberry fruit is rich in bioactive compounds which are beneficial to health. Fruit hybridization can improve quality and metabolite content, but this has yet to be explored in bayberry. Here, we investigated the effects of hybridization on fruit quality and the metabolomic profile of a newly developed hybrid 'BD-107' compared to the parent 'Biqi' and 'Dongkui', the main cultivars in China. Physiochemical evaluations indicated that 'BD-107' outperformed both parental cultivars in crucial quality attributes such as firmness, sugar content, and vitamin C. Metabolomic analysis revealed that hybridization positively influenced the overall metabolite content, with the highest alterations in terpenoids (21.4 %) and flavonoids (12.2 %). Notably, the novel hybrid 'BD-107' contains seven unique metabolites that are not in either parent; Safranal, Myrcenon, p-Menth-3-en-1-ol, Balanophonin B, Swertisin, Genistein-8-C-glucoside, and Ethanone, 1-(1,4-dimethyl-3-cyclohexen-1-yl), linked to antioxidant and health-promoting properties. Our findings provide valuable insights into the influence of bayberry heterosis on terpenoid and flavonoid biosynthesis, enhancing bioactive metabolites and fruit quality, which meet the demand for health-promoting fruit.
Collapse
Affiliation(s)
- Mostafa Saeed
- Institute of Fruit Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China; Department of Pomology, Faculty of Agriculture, Alexandria University, Alexandria 21545, Egypt
| | - Mohamed A Elsadek
- Department of Horticulture, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Zhuyun Chen
- Institute of Fruit Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Lan Zhao
- Institute of Fruit Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Guoyun Wang
- Yuyao Agriculture Technology Extension Center, Ningbo, Yuyao 315400, China
| | - Chaochao Zhou
- Yuyao Agriculture Technology Extension Center, Ningbo, Yuyao 315400, China
| | - Deli Sun
- Agricultural Experiment Station, Zhejiang University, Hangzhou 310058 Zhejiang, China
| | - Zhongshan Gao
- Institute of Fruit Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China.
| | - Yun Jiao
- Institute of Forestry, Ningbo Academy of Agricultural Science, Ningbo 315040, China.
| |
Collapse
|
59
|
Liu X, Xu J, Yu C, Dai C, Chen J, Zhong J, Yang Y, Lin H, Chen X, Zhang Q, Dai L, Zhang J, Zha D, Ye ZC. A novel carbamate-based hybrid derivative with anti-neuroinflammatory properties as a selective butyrylcholinesterase inhibitor for Alzheimer's disease therapy. Bioorg Chem 2025; 161:108551. [PMID: 40347768 DOI: 10.1016/j.bioorg.2025.108551] [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: 01/25/2025] [Revised: 04/22/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
Cholinesterase inhibitors (ChEIs) are widely utilized for the symptomatic management of Alzheimer's disease (AD) by enhancing acetylcholine levels to improve cognitive function. Concurrently, neuroinflammation has emerged as a critical factor in AD progression, necessitating therapies that address this pathology. In this study, we designed and synthesized a novel bifunctional cholinesterase inhibitor, (E)-4-(2-(3-(benzyloxy)-4-oxo-4H-pyran-2-yl) vinyl)-1,2-phenylene bis(ethyl(methyl)carbamate) (D40), which combines potent cholinesterase inhibition with robust anti-neuroinflammatory activity. D40 demonstrated potent inhibition of human butyrylcholinesterase (hBuChE), with an IC₅₀ value of 0.59 ± 0.03 μM, significantly outperforming Rivastigmine (IC₅₀ = 3.70 ± 0.96 μM). Molecular docking and molecular dynamics simulations confirmed a stable and selective binding of D40 to the BuChE active site, underpinning its inhibitory profile. Additionally, D40 exhibited strong anti-inflammatory effects, with an IC₅₀ value of 4.55 ± 0.78 μM for suppressing nitric oxide production and demonstrated excellent blood-brain barrier permeability. In vivo studies in aged 5 × FAD mice revealed that D40 significantly reduced neuroinflammation by suppressing pro-inflammatory cytokines and glial activation. Furthermore, D40 mitigated Aβ deposition, promoted neuronal survival, and improved cognitive deficits, while demonstrating a favorable safety profile in acute toxicity evaluations. These findings highlight D40 as a dual-function ChEI capable of providing symptomatic relief and modulating neuroinflammatory pathways associated with AD. With its enhanced cholinesterase inhibition and anti-inflammatory properties, D40 emerges as a promising candidate for the treatment of advanced stages of AD. Acetylcholine deficiency and neuroinflammation as drivers of Alzheimer's disease dually intervened by Compound D40.
Collapse
Affiliation(s)
- Xueyan Liu
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China; Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jiexin Xu
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Chuanyu Yu
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Chaoxian Dai
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jiajing Chen
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jian Zhong
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Yaoyao Yang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Huiling Lin
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Xili Chen
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Qianling Zhang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Luyao Dai
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jing Zhang
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fujian Province, China.
| | - Daijun Zha
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China.
| | - Zu-Cheng Ye
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China.
| |
Collapse
|
60
|
Priyanka, Rani P, Ranolia A, Kiran, Joshi G, Singh S, Kumar R, Wahajuddin M, Kumar P, Singh D, Sindhu J. Design and synthesis of new 1,4-naphthoquinones appended sulfenylated thiazoles as cyclooxygenase II inhibitors: Exploring the utility in the development of anticancer agents. Bioorg Chem 2025; 161:108537. [PMID: 40334422 DOI: 10.1016/j.bioorg.2025.108537] [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: 01/22/2025] [Revised: 04/05/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
Clinical evaluations revealed the direct role of chronic inflammation in cancer progression. The cyclooxygenase (COX) pathway is particularly important among the various pathways involved in inflammation. There are two COX isoforms: COX-1 and COX-2. The importance of drug discovery lies in selectively inhibiting COX-2, an enzyme expressed during inflammation, unlike COX-1, which is constitutively active. The inhibition of COX-1 is correlated further with gastric ulcers. However, recently approved COX-2 inhibitors have intricate cardiotoxicity, thus creating an utmost need for new COX-2 inhibitors. Considering this, in our present research, we rationally designed and synthesized a series (4a-4q) of 1,4-naphthoquinones appended sulfenylated thiazoles using molecular hybridization approach under metal-free conditions. These synthetics were explored for their anticancer potential against three cell lines, and the hits portraying anticancer effects were further tested against COX isoforms. Among all, compounds 4d and 4f were found to be potent anticancer leads, exhibiting selective inhibition of COX-2 and, concomitantly, the lipoxygenase (LOX) pathway, both of which share the same substrate, arachidonic acid. Both compounds were found to reduce oxidative stress and induce cancer cell death via apoptosis pathway. The experimental outcome was further corroborated using in silico techniques, including density field theory (DFT), Molecular docking, and dynamics.
Collapse
Affiliation(s)
- Priyanka
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Payal Rani
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Anju Ranolia
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Kiran
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, Chauras Campus, HNB Garhwal University (A Central University), Srinagar, Uttarakhand 246174, India; Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, West Yorkshire BD7 1BD, United Kingdom.
| | - Snigdha Singh
- Department of Chemistry, University of Delhi, 110007, India
| | - Roshan Kumar
- Department of Microbiology, Central University of Punjab, VPO-Ghudda, Punjab 151401, India; Department of Microbiology, Graphic Era (Deemed to be University) Clement Town, Dehradun 248002, India
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, West Yorkshire BD7 1BD, United Kingdom
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohtak 124001, India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India.
| |
Collapse
|
61
|
Karthik M, Panchal NK, T M, Bakthavatchalam YD, Neeravi A, Abirami B, Walia K, Veeraraghavan B. Evolutionary insights into NDM variants: Identification and functional analysis of novel NDM-58 in Pseudomonas aeruginosa. Microb Pathog 2025; 204:107574. [PMID: 40228751 DOI: 10.1016/j.micpath.2025.107574] [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: 01/15/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
The emergence of New Delhi Metallo-β-lactamase (NDM) variants in P. aeruginosa has significantly contributed to carbapenem resistance, posing a global threat to antimicrobial therapy. The catalytic activity of NDM, dependent on Zn (II), is enhanced by specific mutations. In this study, we report the identification and characterization of a novel NDM-58 variant (GenBank: OR081828.1) in P. aeruginosa BA24848, which exhibited resistance to multiple β-lactams, including cephalosporins, carbapenems, and BL/BLI combinations. WGS revealed that NDM-58 harbors a unique P185S substitution. This strain is associated with other ARGs (blaPAO, PME-1, fosA, blaOXA-396, blaOXA-494, blaOXA-50, sul1, dfrA1, qacE, aph(3')-VI, qnrVC1, and cat7), indicating a XDR phenotype. Comparative genomic analysis revealed the presence of MGEs (ISpre2, ISPa6, ISPa2, ISPsy29, IS26, Tn4661, ISPa37, and ISUnCu4) associated with NDM-58, which may facilitate the horizontal transfer of resistance determinants. Structural modeling and molecular dynamics simulations demonstrated that NDM-58 exhibits altered stability and compactness compared to NDM-1, likely influencing its enzymatic activity and resistance profile. Residual conservation analysis revealed that Pro185 is highly conserved, and its substitution to serine may impact protein stability and function. The molecular dynamics analysis indicated that NDM-58 has lower residual fluctuations and increased flexibility, which may enhance its adaptability under varying physiological conditions. Our findings provide novel insights into the evolutionary dynamics of NDM enzymes and the role of genetic environments in their dissemination. Understanding these mechanisms is crucial for developing effective surveillance and mitigation strategies against emerging carbapenem-resistant P. aeruginosa.
Collapse
Affiliation(s)
- Maruthan Karthik
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Nagesh Kishan Panchal
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Monisha T
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | | | - Ayyanraj Neeravi
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Baby Abirami
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Kamini Walia
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research, New Delhi, India
| | - Balaji Veeraraghavan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India.
| |
Collapse
|
62
|
Cheng QY, Yang C, Liao YT, Yang SC, Wang HY, Zhou BJ, Li D, Wang WJ, Chen DF, Shao LD. Site-selective structural modification of toosendanin enables one-step synthesis of 12-hydroxyamoorastatin: A natural tautomeric antitumor lead with low toxicity. Bioorg Chem 2025; 161:108578. [PMID: 40367795 DOI: 10.1016/j.bioorg.2025.108578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Natural limonoid triterpenoids toosendanin (1) and 12-hydroxyamoorastatin (5) have been reported to possess broad-spectrum antitumor activity. However, development of antitumor drugs for the treatment of 1 has reached an impasse, due to its severe hepatotoxicity. Notably, compound 5 is a C-12 deacetylated product of 1, but scholars have studied and exploited 5 much less than 1 especially on antitumor activity and toxicity, of which the resource scarcity and structural ambiguity of 5 may be great hindrances. To address these concerns, a site-selective modification of 1 was developed, in which C-12 deacetylation with LiHMDS enabled the one-step synthesis of 5 from 1, solving its insufficient resources. We then revised the 5/7-tautomerization to C-12/C-29-tautomerization of HAR (5) by extensive NMR, LC-MS, DFT-calculations, and X-ray analyses, and determined the relative content of the tautomers in solution phase. Moreover, the biological evaluation of the synthesized derivatives prompted identification of 5 as a natural lead with better antitumor activities in vitro/in vivo. Namely, 5 exhibited antitumor activity through multiple mechanisms, including inhibition of DDR repair, down-regulation of the DNA damage stress-associated transcription factor HSF-1 and its downstream HSPs, promotion of synthetic lethality, disruption of the BAX/BCL2 homeostasis, and elicitation of cellular autophagy. Crucially, 5 achieved 62 % TGI (vs. 17 % for 1) in the JIMT-1 xenograft tumor models without exhibiting hepatotoxicity, providing a solid support to the development of natural limonoid antitumor lead.
Collapse
Affiliation(s)
- Qing-Yan Cheng
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Chen Yang
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yu-Ting Liao
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Shu-Chao Yang
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Hong-Yuan Wang
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Bing-Jie Zhou
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Dashan Li
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Wen-Jing Wang
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Dao-Feng Chen
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China; School of Pharmacy, Institutes of Integrative Medicine, Fudan University, Shanghai 201203, China.
| | - Li-Dong Shao
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
63
|
Vagaggini C, D'Ursi P, Poggialini F, Fossa P, Francesconi V, Trombetti G, Orro A, Dreassi E, Schenone S, Tonelli M, Carbone A. Deciphering the landscape of allosteric glutaminase 1 inhibitors as anticancer agents. Bioorg Chem 2025; 161:108523. [PMID: 40311238 DOI: 10.1016/j.bioorg.2025.108523] [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: 01/17/2025] [Revised: 04/02/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
Glutamine is the second most utilised energy source after glucose for cancer cells to support their proliferation and survival. Glutaminase 1 (GLS1) is the rate-limiting enzyme during the glutaminolysis pathway and thus represents a promising therapeutic target for the development of innovative antitumor agents. Two main classes of GLS1 inhibitors, based on their different binding mode, are reported: the substrate active site and the allosteric site inhibitors. Despite the intense efforts made to date, only two GLS1 inhibitors (i.e.,CB-839 and IPN60090) have entered clinical trials. Therefore, this research field remains to be explored to improve the effectiveness of anticancer therapy. Hence, we describe the discovery and development of reversible allosteric GLS1 inhibitors disclosed in the last six years, dividing them based on their structural similarity with bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) and CB-839. Furthermore, macrocyclic and thiadiazole derivatives, and other structurally different compounds are discussed to present a wider picture of the chemical space under investigation. The study of the binding interactions governing GLS1 inhibition is also analyzed, to help prospectively refine the structural features for greater efficacy. Interestingly, an overview of a new class of irreversible allosteric inhibitors targeting GLS1 Lys320 key residue is provided for the first time. We also summarize the most important biological studies conducted on CB-839 and IPN60090 and their significance for further assessment. The insights garnered from this paper are expected to guide future drug design endeavours toward the identification of novel therapeutics targeting GLS1 to complement and potentially enhance the arsenal of anticancer medications.
Collapse
Affiliation(s)
- Chiara Vagaggini
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Pasqualina D'Ursi
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi 93, 20054 Segrate, Italy
| | - Federica Poggialini
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Paola Fossa
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi 93, 20054 Segrate, Italy; Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy
| | - Valeria Francesconi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy
| | - Gabriele Trombetti
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi 93, 20054 Segrate, Italy
| | - Alessandro Orro
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi 93, 20054 Segrate, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry, and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Silvia Schenone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Michele Tonelli
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy.
| | - Anna Carbone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy.
| |
Collapse
|
64
|
Niu X, Li B, Luo F, Li W, Zhou X, Zhao W. VISTA as a context-dependent immune checkpoint: Implications for tumor immunity and autoimmune pathogenesis. Biochim Biophys Acta Rev Cancer 2025; 1880:189351. [PMID: 40350098 DOI: 10.1016/j.bbcan.2025.189351] [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: 12/18/2024] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
V-domain Ig suppressor of T cell activation (VISTA) is a recently characterized as immune checkpoint regulator with critical roles in modulating immune responses across pathological contexts. In cancer, VISTA contributes to immune evasion by sustaining an immunosuppressive tumor microenvironment, emerging as a promising target for immunotherapeutic intervention. In contrast, in autoimmune diseases, VISTA preserves peripheral immune tolerance and suppresses aberrant immune activation, thereby preventing tissue destruction. This functional dichotomy reflects the complexity of VISTA-mediated signaling, which is modulated by cellular context, microenvironmental cues, and disease stage. Recent studies have elucidated key aspects of VISTA biology, including its structural features, ligand interactions, and context-dependent expression patterns. VISTA operates as a co-inhibitory molecule in cancer, while exerting co-stimulatory or regulatory effects in autoimmunity. This review provides a comprehensive overview of VISTA's discovery, molecular mechanisms, and dual roles in cancer and autoimmune pathogenesis. Furthermore, the current status of VISTA-targeted therapeutic strategies is critically examined, highlighting the translational challenges posed by discrepancies between preclinical models and clinical trial outcomes. Finally, the potential of targeting VISTA within the broader paradigm of immune checkpoint plasticity is discussed, with emphasis on overcoming compensatory immune resistance to enhance therapeutic efficacy. A deeper mechanistic understanding of VISTA is essential for the rational design of future immunomodulatory therapies tailored to specific disease contexts.
Collapse
Affiliation(s)
- Xiaoshuang Niu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Beibei Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Feiyu Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Wanqiong Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xiuman Zhou
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
65
|
Li Y, Du B, Yu L, Luo H, Rong H, Gao X, Yin J. Strategies and challenges of cytosolic delivery of proteins. J Drug Target 2025; 33:837-852. [PMID: 39862226 DOI: 10.1080/1061186x.2025.2458616] [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: 12/09/2024] [Revised: 01/11/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
The cytosolic delivery of therapeutic proteins represents a promising strategy for addressing diseases caused by protein dysfunction. Despite significant advances, efficient delivery remains challenging due to barriers such as cell membrane impermeability, endosomal sequestration and protein instability. This review summarises recent progress in protein delivery systems, including physical, chemical and biological approaches, with a particular focus on strategies that enhance endosomal escape and targeting specificity. We further discuss the clinical translatability of these approaches and propose future directions for improving delivery efficiency and safety, ultimately unlocking the therapeutic potential of intracellular proteins.
Collapse
Affiliation(s)
- Yuanyuan Li
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Baojie Du
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lichao Yu
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hong Luo
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haibo Rong
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Xiangdong Gao
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Yin
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
66
|
Jung JY, Jeong HJ, Han GD. Antimelanogenic effect of fermented licorice water extract on murine melanoma B16F10 cells. Food Sci Biotechnol 2025; 34:2571-2580. [PMID: 40492052 PMCID: PMC12145374 DOI: 10.1007/s10068-025-01878-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 06/11/2025] Open
Abstract
This study investigated the skin-whitening effect of fermented licorice water extract (FLWE) on B16F10 melanocytes. Bioconversion mediated by Issatchenkia orientalis MFST-based fermentation altered licorice's bioactive components. FLWE significantly increased polyphenol and flavonoid content compared with non-fermented licorice water extract (LWE). FLWE more effectively inhibited melanocyte-stimulating hormone-induced melanin production and tyrosinase activity than LWE. RT-PCR and Western blotting revealed that FLWE inhibits the expression of microphthalmia-associated transcription factor (MITF), the transcription factor for TRP-1, TRP-2, and tyrosinase. On analyzing FLWE's inhibitory activity against preexisting tyrosinase within melanocytes, FLWE did not directly inhibit tyrosinase itself, suggesting that FLWE exerts its whitening effect by inhibiting MITF activation. TLC and HPLC indicated that FLWE's superior whitening effect emanates from increased levels of aglycone compounds (isoliquiritigenin, glycyrrhizic acid, licochalcone A, and glabridin) after licorice fermentation. Overall, FLWE is a potentially effective skin-whitening material without side effects (vitiligo) commonly associated with direct tyrosinase inhibition. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-025-01878-z.
Collapse
Affiliation(s)
- Jin Yeong Jung
- Department of Food Science and Technology, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541 Republic of Korea
| | - Ho Jeong Jeong
- Department of Food Science and Technology, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541 Republic of Korea
| | - Gi Dong Han
- Department of Food Science and Technology, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541 Republic of Korea
| |
Collapse
|
67
|
Zhou C, Yang S, Wang J, Pan W, Yao H, Li G, Niu M. Recent advances in PROTAC-based antiviral and antibacterial therapeutics. Bioorg Chem 2025; 160:108437. [PMID: 40215946 DOI: 10.1016/j.bioorg.2025.108437] [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: 01/27/2025] [Revised: 03/05/2025] [Accepted: 04/01/2025] [Indexed: 05/04/2025]
Abstract
By harnessing the ubiquitin proteasome system, proteolysis targeting chimeras (PROTACs) have emerged as a highly promising strategy in drug design for degrading pathogenic proteins. The extensive benefits of PROTAC technology have facilitated its swift and extensive adoption, resulting in numerous PROTACs advancing to clinical trials, and most of them was used for cancers, neurodegenerative diseases, and immune disorders in clinical trials. A number of antiviral PROTACs and antibacterial PROTACs have been developed, exhibiting encouraging bioactivities against various pathogenic viruses and bacterial. Herein, this review summarizes recent advances in PROTAC technology for antiviral and antibacterial drugs, we also provided an overview of the current state of PROTAC clinical trials and detailed the crystal structures of PROTAC in complex with its target protein. Hopefully, this review will contribute to the development of novel antiviral and antibacterial drugs through the utilization of PROTAC technology.
Collapse
Affiliation(s)
- Can Zhou
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen 518000, China
| | - Shiwei Yang
- Shenzhen Second People's Hospital, Shenzhen 518000, China
| | - Jun Wang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Wei Pan
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Hongliang Yao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China
| | - Gang Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China.
| | - Minhong Niu
- Shenzhen Second People's Hospital, Shenzhen 518000, China.
| |
Collapse
|
68
|
Mohamed AR, Georgey HH, Aidy EA, Al-Shafie TA, Elnagar MR, Ali MA, Elblehi SS, Alzahrani AYA, Mousa MHA. Unveiling the potential of xanthines, discovery of potential 7-benzyl-1,3-dimethyl-3,7-dihydro-1H-purine-2,6-dione derivatives with antifibrotic activity for liver injury. Bioorg Chem 2025; 160:108441. [PMID: 40199013 DOI: 10.1016/j.bioorg.2025.108441] [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: 02/27/2025] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
A new series of xanthine-based derivatives were designed, synthesized, and investigated to achieve promising antifibrotic and antioxidant agents for management of liver injury. Structure-based optimizations of the methylxanthine-based KMUP-1 (IX) were performed for inhibiting NF-κB activation pathway. All the newly designed xanthine derivatives 3, 4, 5, 6a-d, 7a-d, and 9a-d were in vitro screened for the antioxidant activity using the DPPH method. Compounds 4 and 5 showed the highest antioxidant activity with an IC50 of 28.02 and 36.02 μM, respectively. Compounds 9c and 9d retained a promising interception of the NF-κB activation pathway in molecular docking simulations within I-κB kinase α (IKKα) crystal structure (PDB ID: 5EBZ). Subsequently, compounds 9c and 9d were evaluated for their in vivo antifibrotic and chemoprotective activity using CCl4-induced hepatic fibrosis rat model. Compounds 9c and 9d successfully ameliorated liver fibrosis, as evidenced by the improved liver histopathological examination and liver enzyme activity levels. Compounds 9c and 9d were evaluated for their effects on mRNA expression levels of key genes involved in liver fibrosis via real-time PCR assays. Compound 9c exhibited a greater inhibitory effect on the expression levels of NF-κB and HIF-1α and a more pronounced stimulation of Nrf2 than compound 9d. Moreover, all the new xanthine derivatives were screened for the cytotoxic activity against the NCI tumor cell lines. Compounds 9c and 9d revealed a non-significant cytotoxic activity against all the assayed tumor cell lines, which indicate their selectivity for the antifibrotic activity. While compounds 6a and 6c displayed promising selective activity against melanoma SK-MEL-5 cell line (GI = 125.6, 90.3 %, respectively), and breast T-47D cell line (GI =87.8, 80.6 %, respectively). The utilized design approach unveiled the versatility of xanthine scaffold to deliver potential antioxidant, liver antifibrotic and chemoprotective agents, along with anticancer candidates via structure modification and optimization.
Collapse
Affiliation(s)
- Abdalla R Mohamed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo 11829, Egypt.
| | - Hanan H Georgey
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Esraa A Aidy
- Medical Biochemistry and Molecular Biology Unit, Cancer Biology Department, National Cancer Institute (NCI), Cairo University, Cairo 11796, Egypt
| | - Tamer A Al-Shafie
- Department of Oral Biology (Biochemistry), Faculty of Dentistry, Pharos University in Alexandria (PUA), Alexandria 21648, Egypt
| | - Mohamed R Elnagar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo 11823, Egypt; Department of Pharmacology, College of Pharmacy, The Islamic University, Najaf 54001, Iraq
| | - Mennatallah A Ali
- Pharmacology and Toxicology Department, PharmD Program, Egypt-Japan University of Science and Technology (E-JUST), Alexandria 21934, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria 22758, Egypt
| | - Abdullah Y A Alzahrani
- Department of Chemistry, Faculty of Science, King Khalid University, Mohail Assir 61421, Saudi Arabia
| | - Mai H A Mousa
- Pharmaceutical Chemistry Department, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo 19346, Egypt
| |
Collapse
|
69
|
Yan F, Sun Y, Zhang S, Jia Y, Zhang J, Huang L, Xu Q, Zhang Y, Chen S, Wu X, Li R. Computer aided design of CGA-N9 derived peptides based on oligopeptide transporters and their antifungal evaluations. Bioorg Chem 2025; 160:108485. [PMID: 40267776 DOI: 10.1016/j.bioorg.2025.108485] [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: 01/24/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
CGA-N9 is an antifungal peptide that primarily targets Candida spp. with a mild activity. Our preceding research confirmed that the CGA-N9 crosses cell membrane with the assistance of C. tropicalis oligopeptide transporter (CtOPT) -1 and - 9. In this study, CGA-N9-derived peptides were designed following the molecular docking results with CtOPT-1 and -9. Compared with CGAN9, they exhibit higher transmembrane efficiency with the assistance of CtOPT-1 during the early phase of transmembrane processes and CtOPT-9 in the late phase. And they displayed significantly enhanced antifungal activity, with lower minimum inhibitory concentrations (MICs) against C. tropicalis, C. albicans, and C. parapsilosis, as well as improved biosafety. Among them, CGAN93 was the most optimizing, with a therapeutic index of 145.33. Furthermore, in a mouse model of systemic candidiasis, CGAN93 demonstrated a therapeutic effect comparable to fluconazole, significantly improving the survival rate of mice, attenuating organ damage, and enhancing the immune organ index. In conclusion, OPTs-based computer aided design is an effective strategy for enhancing the activities of antimicrobial peptides (AMPs) by improving transmembrane transport efficiency. CGAN93 is a promising drug candidate for treating Candidiasis.
Collapse
Affiliation(s)
- Fu Yan
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Yiqing Sun
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Shaojie Zhang
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Yifan Jia
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Jinhua Zhang
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Liang Huang
- School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Qiang Xu
- Zhengzhou Giant Biochemical Group Co., Ltd, 450001 Zhengzhou, Henan, PR China
| | - Yinzhi Zhang
- Zhengzhou Giant Biochemical Group Co., Ltd, 450001 Zhengzhou, Henan, PR China
| | - Shihua Chen
- School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Xingquan Wu
- School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China
| | - Ruifang Li
- Zhengzhou Key Laboratory of Functional Molecules for Biomedical Research, Henan University of Technology, 450001 Zhengzhou, Henan, PR China; School of Biological Engineering, Henan University of Technology, 450001 Zhengzhou, Henan, PR China.
| |
Collapse
|
70
|
Terenteva O, Mostovaya O, Bukharov M, Mukhametzyanov T, Bikmukhametov A, Lyubina A, Voloshina A, Petrov K, Padnya P, Stoikov I. Peptidomimetics based on thiacalixarene with L-tyrosine moieties: Antibacterial activity against methicillin-resistant Staphylococcus aureus and degradation induced by binding to α-chymotrypsin. Bioorg Chem 2025; 160:108434. [PMID: 40187027 DOI: 10.1016/j.bioorg.2025.108434] [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: 02/11/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The design of new antimicrobial agents is an important challenge due to the growing resistance of microorganisms to existing antibiotics. In recent years, the trend towards the development of compounds and materials with (bio)degradable properties has emerged. In this work, we propose and develop a method for the synthesis of new peptidomimetics, i.e., water-soluble macrocyclic quaternary ammonium salts containing L-tyrosine fragments based on p-tert-butylthiacalix[4]arene in various stereoisomeric forms (cone, partial cone, and 1,3-alternate). These compounds have low cytotoxicity (IC50 = 80-267 μM) and high antibacterial activity (MIC = 0.5-15.6 μM) against Gram-positive bacterial strains including methicillin-resistant Staphylococcus aureus (MRSA). The obtained peptidomimetics can bind α-chymotrypsin with the formation of supramolecular systems and their subsequent degradation. Our results demonstrate the first example of multi-action thiacalixarene derivatives with antibacterial activity, protein binding ability and degradation induced by binding to α-chymotrypsin. The obtained results open the possibility of creating multi-action peptidomimetic systems with antimicrobial and biodegradable effect.
Collapse
Affiliation(s)
- Olga Terenteva
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Olga Mostovaya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Mikhail Bukharov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Timur Mukhametzyanov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Azamat Bikmukhametov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation
| | - Anna Lyubina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Alexandra Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Konstantin Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 8 Arbuzov Street, Kazan 420088, Russian Federation
| | - Pavel Padnya
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation.
| | - Ivan Stoikov
- A.M. Butlerov Chemical Institute, Kazan Federal University, 18 Kremlevskaya Street, Kazan 420008, Russian Federation.
| |
Collapse
|
71
|
Eldehna WM, Tawfik HO, Nafie MS, Al Kamaly O, El-Hamaky AA, El Hassab MA, Elsayed ZM, Elnaggar YSR, Al-Karmalawy AA, di Giacomo V, Balaha M. Novel benzofuran-conjugated indolin-2-ones as anticancer agents; design, synthesis, biological assessments, and molecular modeling insights. Bioorg Chem 2025; 160:108494. [PMID: 40286528 DOI: 10.1016/j.bioorg.2025.108494] [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: 01/28/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors have been authorized for the treatment of breast cancer (BC) and prostate cancer (PC). Recent studies suggest that inhibiting angiogenesis through the vascular endothelial growth factor receptor (VEGFR) enhances cellular sensitivity to PARP inhibitors. This study presents the design, synthesis and full characterization of dual VEGFR-2 and PARP-1 inhibitors obtained by conjugating a PARP-1 inhibitor with VEGFR-2 inhibitor fragments (indole, benzofuran, and piperazine). Four compounds exhibited significant inhibitory activities against human prostate cancer cell lines (PC3) and breast cancer cell lines (MCF7) at 48 h. These compounds were identified as dual VEGFR-2 and PARP-1 inhibitors with low or sub-micromolar ranges, especially 12f, with IC50 values of 0.43 μM and 1.10 μM, respectively. Moreover, the potent compound 12f markedly decreased scratch wound closure and colony formation. Moreover, compound 12f significantly induced apoptosis in PC3 cells and arrested cells at the S phase. The dual inhibition of VEGFR-2 and PARP-1 protein kinase was further validated using western blotting. Applying molecular docking and dynamics determined the target compound's binding mechanism.
Collapse
Affiliation(s)
- Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt.
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Omkulthom Al Kamaly
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Anwar A El-Hamaky
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Mahmoud A El Hassab
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, Egypt
| | - Zainab M Elsayed
- Scientific Research and Innovation Support Unit, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Yosra S R Elnaggar
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Egypt; Head of Research & International Publishing Administration (RIPA), Pharos University in Alexandria, Egypt
| | - 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
| | - Viviana di Giacomo
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marwa Balaha
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt; Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
72
|
de Araújo MRB, da Silveira E Santos DM, da Silva Sousa J, do Prado GF, de Carvalho Castro DL, Soares Dias Júnior M, Perini HF, Minafra C, de Castro Soares S, Rodrigues EB, Lana LF, Gonçalves LAC, Andrade BS, Gomes T, Azevedo V. Systemic Infection Caused by Mycobacterium fortuitum Following Aortic Valve Transplantation-Employing Combined Molecular Techniques for Accurate Species Identification. Curr Microbiol 2025; 82:335. [PMID: 40515850 DOI: 10.1007/s00284-025-04313-y] [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: 10/22/2024] [Accepted: 05/15/2025] [Indexed: 06/16/2025]
Abstract
Mycobacterium fortuitum, a nontuberculous mycobacterium (NTM), is known for causing opportunistic infections, particularly in immunocompromised individuals. Identification of NTM species requires precise diagnostic techniques. Here, we present a study of a systemic infection by M. fortuitum in an elderly patient, identified through MALDI-TOF MS and confirmed by gene sequencing. An 88-year-old Italian man developed an abscess at his pacemaker site, leading to acute infectious endocarditis caused by Staphylococcus capitis. During hospitalization, he experienced multiple complications, including jugular vein thrombosis, pulmonary thromboembolism, and gastrointestinal bleeding. Despite treatment with vancomycin, he developed bacteremia, allowing the isolation of M. fortuitum from blood culture. The strain was identified using MALDI-TOF MS and confirmed by Sanger sequencing of the 16S rRNA and hsp65 genes. Phylogenetic analysis supported the identification. The patient, after treatment with carbapenems and aminoglycosides, returned to Italy without further clinical complications. This case underscores the necessity of precise diagnostic tools for managing NTM infections. The combined application of MALDI-TOF MS and gene sequencing enabled accurate species identification, which is essential for guiding effective treatment of systemic M. fortuitum infections, especially in complex clinical scenarios involving immunosuppression.
Collapse
Affiliation(s)
- Max Roberto Batista de Araújo
- Operational Technical Nucleus, Microbiology, Hermes Pardini Institute (Fleury Group), Vespasiano, Minas Gerais, Brazil.
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | | | - Jailan da Silva Sousa
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Diogo Luiz de Carvalho Castro
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Moacir Soares Dias Júnior
- Molecular Biology, Operational Technical Nucleus, Hermes Pardini Institute (Fleury Group), Vespasiano, Minas Gerais, Brazil
| | - Hugo Felix Perini
- Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Cíntia Minafra
- School of Veterinary and Animal Science, Food Research Center, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Siomar de Castro Soares
- Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Evandro Bento Rodrigues
- Operational Technical Nucleus, Microbiology, Hermes Pardini Institute (Fleury Group), Vespasiano, Minas Gerais, Brazil
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luan Freitas Lana
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Bruno Silva Andrade
- Laboratory of Bioinformatics and Computational Chemistry, State University of Southwest Bahia, Jequié, Bahia, Brazil
- INRAE, Institut Agro, STLO, Rennes, France
| | - Talita Gomes
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vasco Azevedo
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
73
|
Shrivastava D, Gupta LK, Yerrapureddy AR, Nune Satya SK. Discovery of novel compound IGYZT01060 for inhibition of IRAK-4 enzyme for the treatment of sepsis. Biotechnol Lett 2025; 47:62. [PMID: 40515955 DOI: 10.1007/s10529-025-03604-5] [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: 11/26/2024] [Revised: 05/19/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025]
Abstract
PURPOSE This study aimed at the discovery of a novel chemical entity, IGYZT01060 for inhibiting IRAK-4 (Interleukin-1 receptor-associated kinase 4) and testing its ability to inhibit the IRAK-4 enzyme, that is crucial in the context of sepsis due to its central role in the innate immune response, particularly in mediating inflammatory signals from Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1Rs). METHODS This study explores the synthesis and characterization of a novel compound, IGYZT01060, designed to inhibit IRAK-4. The inhibitory analysis was carried out in silico, in vitro and in vivo. Molecular docking was done using AutoDock software. The in vitro assays were carried out in LPS induced THP-1 cells. ADME assays were carried out for understanding the drug delivery and distribution pattern. Lastly, in vivo mice sepsis model using intraperitoneally administered LPS and orally administered compound IGYZT01060 was studied estimate the efficacy of compound IGYZT01060. RESULTS In silico docking analyses demonstrated a high affinity of IGYZT01060 for IRAK-4, with target prediction indicating a preference for kinase enzymes. The NMR spectroscopy confirmed the successful synthesis and purity of the compound. In vitro studies revealed that IGYZT01060 effectively inhibits IRAK-4 with an IC50 of less than 100 nM. Pharmacokinetic evaluations indicated a favorable clearance rate and high bioavailability, essential for the therapeutic efficacy of any drug. Furthermore, the mice sepsis model results indicated a significant inhibition of IRAK-4, almost as good as the corticosteroid dexamethasone. CONCLUSION The promising IRAK-4 inhibition demonstrated by our compound IGYZT01060, along with its favourable pharmacokinetic profile and significant efficacy in a mice sepsis model, highlights its potential as a powerful therapeutic option for treating inflammatory conditions.
Collapse
Affiliation(s)
- Deepali Shrivastava
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur, Andhra Pradesh, 522213, India
| | - Lavleen K Gupta
- IgY Immunologix India Pvt Ltd, 3-14/2, Narsingi, Hyderabad, 500089, India
| | | | - Sampath Kumar Nune Satya
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur, Andhra Pradesh, 522213, India.
| |
Collapse
|
74
|
Fan XC, Wang J. Hederagenin promotes SIRT6 to attenuate epidural scar formation by aggravating PRMT1 deacetylation. Bone Joint Res 2025; 14:516-526. [PMID: 40511498 PMCID: PMC12163784 DOI: 10.1302/2046-3758.146.bjr-2024-0287.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/16/2025] Open
Abstract
Aims The formation of a postoperative epidural scar induced by epidural fibrosis is the main reason for recurrence of lumbar disc herniation after laminectomy. Hederagenin (HE) has been found to be widely present in various medicinal plants and has various pharmacological functions. This study aimed to investigate the effect and regulatory mechanism of HE on epidural scar formation. Methods Transforming growth factor beta 1 (TGF-β1)-stimulated epidural scar fibroblasts were used as an in vitro cell model. Based on that, HE treatment was carried out along with sirtuin-6 (SIRT6) silence or protein arginine N-methyltransferase 1 (PRMT1) overexpression. The interaction between SIRT6 and PRMT1 was evaluated by pulldown and co-immunoprecipitation (CoIP) assays. Then, cell proliferation, apoptosis, and fibrosis were measured by Cell Counting Kit (CCK)-8, flow cytometry, and western blotting. Moreover, the effects of receptor activator of nuclear factor-κB ligand (RANKL) supplementation and endoplasmic reticulum (ER) stress were also evaluated by supplementing recombinant protein and specific inhibitor or activator. Results HE depressed cell proliferation and fibrosis, while inducing apoptosis of epidural fibroblasts. Meanwhile, HE promoted SIRT6 expression which suppressed PRMT1 acetylation and protein stability. Additionally, HE induced ER stress and upregulated RANKL in epidural fibroblasts via mediating SIRT6/PRMT1 axis. Conclusion Generally, the therapeutic role of HE treatment on epidural scar formation was exerted by regulating SIRT6/PRMT1 axis-mediated ER stress and RANKL pathway. This study provides evidence of a novel therapeutic measure for epidural scar formation.
Collapse
Affiliation(s)
- Xiao-Chen Fan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiao Tong University, Xi'an, China
- Department of Traditional Chinese Medicine, Honghui Hospital, Xi’an, China
| | - Jue Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiao Tong University, Xi'an, China
| |
Collapse
|
75
|
Han R, Balsiger T, Dobrzyński M, Dürr L, Hell T, Smieško M, Solis PN, Hamburger M, Pertz O, Teufel R, Garo E. Coumarins and betulinic acid analogues from Mammea americana and their inhibitory activities on oncogenic MAPK/ERK and PI3K/AKT pathways in human melanoma cells. PHYTOCHEMISTRY 2025:114580. [PMID: 40516889 DOI: 10.1016/j.phytochem.2025.114580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 06/11/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
Melanoma, a highly aggressive form of skin cancer, is primarily driven by two key oncogenic signaling pathways: MAPK/ERK and PI3K/AKT. In a discovery program aiming to identify natural products that inhibit one or both pathways, an in-house library of 2'576 plant extracts was screened using a high-content screening assay with melanoma cells expressing ERK/AKT activity biosensors to quantify inhibition at the single-cell level. The ethyl acetate extract from the leaves of Mammea americana was found to inhibit both pathways in the patient-derived cell line MM121224 and was selected for HPLC-based activity profiling. Scale-up isolation of the compounds eluting in the active window of the chromatogram afforded ten previously undescribed betulinic acid analogues (1-10), along with nine known coumarins (11-19). The isolated compounds were individually evaluated for their inhibitory activity on ERK and AKT in MM121224 cells. Interestingly, none of the betulinic acid derivatives 1-10 showed activity in the assay. In contrast, the isolated coumarins were shown to inhibit both pathways, with the most potent theraphin B (11) exhibiting an IC50 value of 37.0 ± 0.4 μM against the AKT pathway, while also demonstrating weaker activity against the ERK pathway.
Collapse
Affiliation(s)
- Ruzhe Han
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Tamara Balsiger
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Maciej Dobrzyński
- Institute of Cell Biology, University of Bern, Bern, 3012, Switzerland
| | - Lara Dürr
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Tanja Hell
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Martin Smieško
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Pablo N Solis
- Centro de Investigaciones Farmacognosticas de la Flora Panameña - Dr. Mahabir P. Gupta, Faculty of Pharmacy, University of Panama, Panama City, 3366-4, Republic of Panama
| | - Matthias Hamburger
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Olivier Pertz
- Institute of Cell Biology, University of Bern, Bern, 3012, Switzerland
| | - Robin Teufel
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland
| | - Eliane Garo
- Department of Pharmaceutical Sciences, University of Basel, Basel, 4056, Switzerland.
| |
Collapse
|
76
|
Beck E, Bröllos D, Krummenacher I, Kupfer T, Dietz M, Wellnitz T, Mihm C, Braunschweig H. Boron-chalcogen heterocycles and linear tetraboranes from a cyclic tetra(amino)tetraborane. Nat Commun 2025; 16:5304. [PMID: 40506450 PMCID: PMC12163077 DOI: 10.1038/s41467-025-60549-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
While small carbocyclic rings have long been recognized as pivotal building blocks in chemistry, their all-boron counterparts have remained largely unexplored. In this work, we present a detailed account of the functionalization reactivity of our cyclic tetraborane B4(NCy2)4 (Cy = cyclohexyl) encompassing both ring-expansion and ring-opening reactions. Specifically, diphenyl dichalcogenides effect ring expansion to five-membered B4E rings (E = S, Se, Te), while halogenating agents induce ring opening to generate linear tetraboranes with halide end groups. These transformations reveal reactivity patterns reminiscent of strained organic ring systems, thus highlighting the cyclic tetraborane's potential as a versatile precursor for synthesizing intricate boron-rich architectures.
Collapse
Affiliation(s)
- Eva Beck
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
- Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Diana Bröllos
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
- Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Ivo Krummenacher
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
- Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Thomas Kupfer
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
- Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Maximilian Dietz
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
- Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Tim Wellnitz
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
- Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Cornelius Mihm
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
- Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany
| | - Holger Braunschweig
- Institut für Anorganische Chemie, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany.
- Institute for Sustainable Chemistry & Catalysis with Boron, Julius-Maximilians-Universität Würzburg, Am Hubland, Würzburg, Germany.
| |
Collapse
|
77
|
Chisholm TS, Hunter CA. Ligands for Protein Fibrils of Amyloid-β, α-Synuclein, and Tau. Chem Rev 2025; 125:5282-5348. [PMID: 40327808 PMCID: PMC12164286 DOI: 10.1021/acs.chemrev.4c00838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 05/08/2025]
Abstract
Amyloid fibrils are characteristic features of many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. The use of small molecule ligands that bind to amyloid fibrils underpins both fundamental research aiming to better understand the pathology of neurodegenerative disease, and clinical research aiming to develop diagnostic tools for these diseases. To date, a large number of amyloid-binding ligands have been reported in the literature, predominantly targeting protein fibrils composed of amyloid-β (Aβ), tau, and α-synuclein (αSyn) fibrils. Fibrils formed by a particular protein can adopt a range of possible morphologies, but protein fibrils formed in vivo possess disease-specific morphologies, highlighting the need for morphology-specific amyloid-binding ligands. This review details the morphologies of Aβ, tau, and αSyn fibril polymorphs that have been reported as a result of structural work and describes a database of amyloid-binding ligands containing 4,288 binding measurements for 2,404 unique compounds targeting Aβ, tau, or αSyn fibrils.
Collapse
Affiliation(s)
- Timothy S. Chisholm
- Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.
| | - Christopher A. Hunter
- Yusuf Hamied Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.
| |
Collapse
|
78
|
Coppola G, Cennamo F, Ciccone G, Ibba ML, Ruscio AD, Vito AD, Esposito CL, Catuogno S. Aptamer-based applications in delivering cancer gene therapies and beyond: state of the art and the missing links to clinical translation. Adv Drug Deliv Rev 2025:115639. [PMID: 40513886 DOI: 10.1016/j.addr.2025.115639] [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: 04/11/2025] [Revised: 06/03/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
The possibility of correcting genetic and epigenetic alterations through gene therapies has been considered a cornerstone in oncology. However, modest results have been achieved in clinics, mainly due to inefficient tumor targeting and side effects. Nucleic acid aptamers are three-dimensional folded single-stranded DNAs or RNAs that selectively bind receptors on cellular membranes, being subsequently internalized via receptor-mediated endocytosis. Thanks to this capability, internalizing aptamers have been investigated as targeting moieties to deliver gene therapies more efficiently and selectively in tumor cells. Promising preclinical results suggested that aptamers could represent the long-awaited step forward in cancer gene therapy. Nevertheless, no clinical trials of aptamer-based gene therapies have been carried out two decades after the first preclinical application, indicating the field could not be sufficiently mature for translatability. The review aims to update thestate of the art regarding aptamers' contribution to gene therapy delivery and to critically highlight the main shortcomings that could have hindered clinical evaluations. In addition, pioneering insights regarding the use of aptamers as co-factors in CRISPR/Cas9 technology or as direct epigenetic regulators are also summarized, revealing more extended applicability not limited to the delivery of cancer gene therapies.
Collapse
Affiliation(s)
- Gabriele Coppola
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Fabiola Cennamo
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Giuseppe Ciccone
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", 81100 Caserta, Italy.
| | - Maria Luigia Ibba
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy; Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Caserta, Italy.
| | - Annalisa Di Ruscio
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA; Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Boston, MA 02215, USA.
| | - Aldo Di Vito
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Carla Lucia Esposito
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Silvia Catuogno
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| |
Collapse
|
79
|
Bartz FM, Sałat K, Urbańska K, Lemke J, Rosendahl P, Schmidt L, Schulig L, Garscha U, Link A, Bednarski PJ. New K V7.2/3 Channel Activators Exhibit Superior Toxicity and Metabolic Profiles to Flupirtine and Demonstrate Promising In Vivo Analgesic Effects. ACS Chem Neurosci 2025. [PMID: 40492583 DOI: 10.1021/acschemneuro.5c00278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025] Open
Abstract
The first-in-class KV7.2/3 channel activator flupirtine, was considered a potent analgesic in various pain conditions. However, it was withdrawn from the market in 2018 due to severe hepatotoxicity associated with forming reactive metabolites. In this work, we present new KV7.2/3 channel modulators that have been evaluated in several preclinical mouse pain models, including acute thermally and chemically induced pain, diabetes-induced neuropathic pain, and chemotherapy-induced peripheral neuropathy. In addition, the new KV7.2/3 channel activators were compared with the reference substances flupirtine, retigabine, and azetukalner, focusing on the inhibition of the hERG channel, nephrotoxicity, metabolic stability, and the formation of reactive metabolites. A flupirtine analog with a pyrimidine scaffold (8) showed clear advantages over the reference compounds tested, with a favorable toxicity profile, a 2 h in vitro half-life when incubated with human liver microsomes, and a 9-fold reduction in the formation of reactive metabolites compared to flupirtine. This compound also demonstrated strong in vivo efficacy in pain models, making it a promising candidate for further development of KV7.2/3 channel activators.
Collapse
Affiliation(s)
- Frieda-Marie Bartz
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Kinga Sałat
- Faculty of Pharmacy, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Katarzyna Urbańska
- Faculty of Pharmacy, Chair of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Jana Lemke
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Pascal Rosendahl
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Louis Schmidt
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Lukas Schulig
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Ulrike Garscha
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Andreas Link
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Patrick J Bednarski
- Institute of Pharmacy, Department of Pharmaceutical and Medicinal Chemistry, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| |
Collapse
|
80
|
Dai X, Jiang D, Gou C, Mei K, Sun B, Liu J, Yao N, Liu X, Qiu N. Biotinylated Self-Assembled Cytarabine Nano-Prodrug with Enhanced Targetability and Anticancer Efficacy: An In Vitro Study. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:14197-14210. [PMID: 40450601 DOI: 10.1021/acs.langmuir.5c01147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
In this study, we developed a tumor-target biotinylated cytarabine (Ara-C) prodrug linked through a disulfide bond (Bio-SS-Ara). To enhance its delivery, Bio-SS-Ara was found to be able to self-assemble into uniform nanoparticles (BSSA NPs) with an average particle size of 230 nm, a PDI of 0.265, a critical micelle concentration (CMC) of 5.89 μg/mL, and a zeta potential of -15 mV. BSSA NPs showed good storage stability over 6 weeks. In a glutathione-rich environment, the drug release of BSSA NPs was more rapid than that of BCCA NPs. Concentration-dependent size analysis suggested that BSSA NPs were formed through a dynamic, reversible assembly process. Cellular uptake studies showed significantly increased internalization of BSSA NPs compared to free Ara-C, with enhancements of 4.13-fold in HeLa cells and 3.13-fold in A549 cells. Cytotoxicity assays demonstrated enhanced efficacy, reducing the IC50 by 67.9% in HeLa and 45.2% in A549 cells. The migration rate of HeLa cells was reduced to 14% after BSSA NPs treatment, in comparison with Ara-C treatment, and nuclear shrinkage and fragmentation were observed, indicating the potential of BSSA NPs to limit tumor progression and induce apoptosis. Importantly, no hemolysis was observed in BSSA NPs-treated groups, confirming their good biocompatibility. These findings suggest that BSSA NPs represent a promising strategy for targeted cancer therapy, combining enhanced drug delivery, antitumor efficacy, and biocompatibility.
Collapse
Affiliation(s)
- Xiaoshuang Dai
- College of Ecology and Environment, Chengdu University of Technology, Chengdu 610059, China
| | - Daobin Jiang
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Chunlin Gou
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Ke Mei
- College of Ecology and Environment, Chengdu University of Technology, Chengdu 610059, China
| | - Bin Sun
- College of Ecology and Environment, Chengdu University of Technology, Chengdu 610059, China
| | - Jianpeng Liu
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Na Yao
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Xinyu Liu
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| | - Neng Qiu
- College of Materials and Chemistry & Chemical Engineering, Chengdu University of Technology, Chengdu 610059, China
| |
Collapse
|
81
|
Narayan S, Nagpal K, Kumar P. Polyphenol-conjugated polysaccharide nanoplatforms for enhanced therapeutic efficacy. Expert Opin Drug Deliv 2025:1-19. [PMID: 40455588 DOI: 10.1080/17425247.2025.2514714] [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: 03/04/2025] [Accepted: 05/29/2025] [Indexed: 06/11/2025]
Abstract
INTRODUCTION Polyphenols represent a broad class of natural chemical compounds comprising, but not limited to, tannins, phenolic acids, flavonoids, flavanones, flavanols, anthocyanins, and their related polymerized derivatives. Polyphenols are an important component of various commercial, naturally derived products with therapeutic properties. However, their full therapeutic potential is restricted by inherently low solubility and limited dispersibility in the aqueous phase. AREAS COVERED This special report provides a focused viewpoint of various polyphenols conjugated with polysaccharides such as chitosan, dextran, curdlan, alginate, gellan, and pectin. The advantages and performance of conjugating polysaccharides to polyphenols are presented and discussed. Further to this, nanoplatforms of polyphenol-conjugated polysaccharides with enhanced therapeutic efficacy and physicochemical properties are discussed. EXPERT OPINION Conjugation of polyphenols with polysaccharides, using various chemical conjugation techniques, may provide an amenable solution to the envisaged polyphenol efficacy challenge. The conjugation of polyphenols with polysaccharides offers multiple advantages, including improved aqueous solubility, enhanced protection against oxidative degradation, and targeted delivery through ligand-functionalized nanocarriers. This approach not only improves the pharmacokinetic profile of polyphenols but also maximizes their therapeutic efficacy while minimizing off-target toxicity. Furthermore, polysaccharide-polyphenol conjugates hold immense potential in functional foods, nutraceuticals, and pharmaceutical formulations, where enhanced bioactivity and controlled release are desired.
Collapse
Affiliation(s)
- Somesh Narayan
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Kalpana Nagpal
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
82
|
Ling H, Xiao S, Lei Y, Zhou Y, Tan J, Chen X, Ma D, Liang C, Liu Q, Liu W, Zeng T. The advancement of ubiquitination regulation in apoptosis, ferroptosis, autophagy, drug resistance and treatment of cancer. Arch Biochem Biophys 2025; 771:110497. [PMID: 40499632 DOI: 10.1016/j.abb.2025.110497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 05/12/2025] [Accepted: 06/01/2025] [Indexed: 06/16/2025]
Abstract
Ubiquitination, a crucial post-translational modification, significantly influences cancer initiation and progression. This review emphasizes its roles in programmed cell death (including apoptosis, ferroptosis, and autophagy), drug resistance, and cancer therapy. In cell death pathways, ubiquitination through K48 and K63 linkages regulates proteins such as Bcl-2, ACSL4, and p62, thereby affecting cancer cell survival. The dysregulation of ubiquitin-specific proteases (USPs), such as USP1 and USP22, leads to uncontrolled cell cycle progression and abnormal DNA repair, which promotes tumorigenesis. In the context of drug resistance, ubiquitination modifies ABC transporters and DNA repair enzymes, facilitating chemotherapy resistance. Additionally, the inhibition of ferroptosis and autophagy-related ubiquitination allows cancer cells to evade apoptosis. In immunotherapy, ubiquitination plays a role in the degradation of PD-1 and PD-L1, as well as in antigen presentation, thereby shaping the immune microenvironment. Therapeutic strategies, including proteasome inhibitors, E3 ligase inhibitors, and PROTACs show promise. Targeting USPs or employing stress-responsive PROTACs may help overcome resistance, with combination therapies emerging as a key area of research. Future studies should aim to clarify the dynamics of the ubiquitination network, develop selective inhibitors, and explore precision medicine for clinical applications.
Collapse
Affiliation(s)
- Hui Ling
- Cancer Research Institute, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Shuyao Xiao
- Cancer Research Institute, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yalin Lei
- Cancer Research Institute, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yujie Zhou
- 2022 Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiaying Tan
- 2021 Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xiaojie Chen
- 2021 Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Da Ma
- 2021 Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Can Liang
- 2021 Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Qingyi Liu
- 2022 Department of Oral Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Wei Liu
- 2022 Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Tiebing Zeng
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
83
|
Ahmed TE, Henary M. Dimerizing Heptamethine Cyanine Fluorophores from the Meso Position: Synthesis, Optical Properties, and Metal Sensing Studies. Org Lett 2025. [PMID: 40490423 DOI: 10.1021/acs.orglett.5c01620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
A series of eight dimeric heptamethine cyanine fluorophores were synthesized, purified, and produced in moderate yields by connecting two heptamethine dyes from their meso position. Their physicochemical properties were predicted, and their optical properties and photothermal stability were studied. They showed selective absorbance and fluorescence quenching in the presence of copper(II) ions. This study serves as a basis for exploring the effects of dimerization of heptamethine cyanine fluorophores and their potential use for various biomedical applications.
Collapse
Affiliation(s)
- Tarek Erfan Ahmed
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Maged Henary
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
- Center For Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30303, United States
| |
Collapse
|
84
|
Elrashedy A, Ibrahim NE, Abo-Salem H, Elaasser MM, El-Sawy ER. Design, synthesis, and molecular modeling of new 1,2,4-triazole-containing indole compounds as aromatase antagonists for the treatment of breast cancer. Bioorg Chem 2025; 163:108677. [PMID: 40489920 DOI: 10.1016/j.bioorg.2025.108677] [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: 04/23/2025] [Revised: 06/01/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
Recently, the incidence of breast cancer has increased among postmenopausal women. This highlights the need for improved aromatase inhibitors with fewer side effects than those currently available. This study focuses on designing and evaluating potential aromatase inhibitors containing the 1,2,4-triazole/indole hybrid, given their proven impact in this respect. Accordingly, by exploiting the thiol and amine groups in 5-((1H-indole-3-yl)methyl)-4-amino-4H-1,2,4-triazole-3-thiol (2) to react with some reagents, new hybrid 1,2,4-triazole-indole compounds were obtained. A cytotoxicity (MTT) test was conducted to assess the anticancer activity of the newly synthesized compounds on the breast cancer cell line MCF-7. The results of the MTT assay indicated that compounds 4d and 5 exhibited significant cytotoxic activity against the MCF-7 cancer cell lines with IC50 values of 17.67 ± 0.34 and 17.01 ± 0.53 μM (respectively) compared with Cisplatin, which showed an IC50 value of 18.03 ± 0.71 μM. Molecular dynamics simulations were conducted to investigate the crystal structure of human placental aromatase cytochrome (AR) P450 for compounds 4d and 5. The results revealed that compound 5 exhibited significant interactions with key residues in the binding site, leading to a stable complex throughout the entire simulation. Depending on molecular dynamics data, compound 5 was selected for an in vitro aromatase inhibition study. Compound 5 inhibited aromatase with an IC50 value of 0.026 μM, which is approximately equivalent to the reference compound letrozole (IC50 0.024 μM).
Collapse
Affiliation(s)
- Ahmed Elrashedy
- Department of the Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Institute, National Research Centre (NRC), 33 El-Behouth St., Dokki, Giza 12622, Egypt; Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Menoufia 32897, Egypt.
| | - Noha E Ibrahim
- Department of Microbial Biotechnology, Biotechnology Research Institute, National Research Centre, 33 El-Behouth St. (former El-Tahrir St.), Dokki, Giza P.O.12622, Egypt
| | - Heba Abo-Salem
- Chemistry of Natural Compounds Department, National Research Centre, 33 El-Behouth St. (former El-Tahrir St.), Dokki, Giza P.O.12622, Egypt
| | - Mahmoud Mohamed Elaasser
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Nasr City, Cairo, Egypt.
| | - Eslam R El-Sawy
- Chemistry of Natural Compounds Department, National Research Centre, 33 El-Behouth St. (former El-Tahrir St.), Dokki, Giza P.O.12622, Egypt.
| |
Collapse
|
85
|
Lepori I, Jackson K, Liu Z, Chordia MD, Wong M, Rivera SL, Roncetti M, Poliseno L, Freundlich JS, Pires MM, Siegrist MS. The Mycomembrane Differentially and Heterogeneously Restricts Antibiotic Permeation. ACS Infect Dis 2025. [PMID: 40471697 DOI: 10.1021/acsinfecdis.4c01062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The recalcitrance of Mycobacterium tuberculosis to antibiotic treatment has been broadly attributed to the impermeability of the organism's outer mycomembrane. However, the studies that support this inference have been indirect or reliant on bulk population measurements. We previously developed the Peptidoglycan Accessibility Click-Mediated AssessmeNt (PAC-MAN) method to covalently trap azide-modified small molecules in the peptidoglycan cell wall of live mycobacteria after they have traversed the mycomembrane. Using PAC-MAN, we now show that the mycomembrane differentially restricts access to fluorophores and antibiotic derivatives. Mycomembranes of both M. tuberculosis and the model organism Mycobacterium smegmatis discriminate between divergent classes of antibiotics as well as between antibiotics within a single family, the fluoroquinolones. By analyzing subpopulations of M. tuberculosis and M. smegmatis, we also found that some fluorophores and vancomycin are heterogeneously restricted by the mycomembrane. Our data indicate that the mycomembrane is a molecule- and cell-specific barrier to antibiotic permeation.
Collapse
Affiliation(s)
- Irene Lepori
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Kiserian Jackson
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Zichen Liu
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Mahendra D Chordia
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Mitchell Wong
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Sylvia L Rivera
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Marta Roncetti
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
- University of Siena, Siena 53100, Italy
| | - Laura Poliseno
- Institute of Clinical Physiology, National Research Council (CNR-IFC), Pisa 56124, Italy
- Oncogenomics Unit, Core Research Laboratory, ISPRO, Pisa 56124, Italy
| | - Joel S Freundlich
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Marcos M Pires
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
- Department of Microbiology, Immunology, and Cancer, University of Virginia, Charlottesville, Virginia 22904, United States
| | - M Sloan Siegrist
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
86
|
Kim YG, Jeon H, Boya BR, Lee JH, Lee J. Targeting biofilm formation in Candida albicans with halogenated pyrrolopyrimidine derivatives. Eur J Med Chem 2025; 290:117528. [PMID: 40121868 DOI: 10.1016/j.ejmech.2025.117528] [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: 01/13/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Growing concern over environmental contaminants, including pharmaceuticals and antifungal agents, highlights their role in promoting resistance and biofilm formation by microorganisms. Antifungal resistance, especially in drug-resistant Candida spp., poses a global threat, worsened by the widespread use of antifungal agents in both clinical applications and environmental contamination. This study investigates the antibiofilm properties of various halogenated pyrrolo pyrimidine derivatives, specifically 4-chloro-5-iodo-7H-pyrrolo[2,3-d]pyrimidine (10) and 2,4-dichloro-5-iodo-7H-pyrrolo[2,3-d]pyrimidine (16), against fluconazole-resistant C. albicans. Both compounds demonstrated strong biofilm inhibition, with 16 showing greater efficacy even at lower concentrations. qRT-PCR analysis revealed downregulation of key biofilm- and hyphae/germ tube-relating genes, including ALS3, HWP1, and ECE1, alongside upregulation of stress response and biofilm regulator genes such as CDR11, GST3, IFD6, UCF1, YWP1, and ZAP1, indicating complex regulatory responses to the treatments. Molecular docking analysis revealed that these compounds bind effectively to the binding cavity of the ALS3 protein, with halogen atoms playing a key role in stabilizing interaction. Compound 16 exhibited minimal cytotoxicity in Brassica rapa and Caenorhabditis elegans models, suggesting a favorable ADMET safety profile. Confocal microscopy analysis confirmed the compounds effectiveness in preventing biofilm formation when applied as biodegradable PLGA coatings on biomaterial surfaces. These findings suggest that 16 holds promise as a potent antifungal agent with reduced environmental impact, offering both efficacy and sustainability.
Collapse
Affiliation(s)
- Yong-Guy Kim
- The Institute of Clean Technology, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea
| | - Hyejin Jeon
- School of Chemical Engineering, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea
| | - Bharath Reddy Boya
- School of Chemical Engineering, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea
| | - Jin-Hyung Lee
- The Institute of Clean Technology, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea.
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehakro, Gyeongsansi, Gyeonsanggbukdo, 38541, Republic of Korea.
| |
Collapse
|
87
|
Ying TT, Hu HQ, Wu XW, Xu XL, Lv J, Zhang SN, Wang H, Hou W, Wei B, Rao GW. Optimized ebselen derivatives as novel potent Escherichia coli β-glucuronidase covalent allosteric inhibitors. Eur J Med Chem 2025; 290:117571. [PMID: 40168911 DOI: 10.1016/j.ejmech.2025.117571] [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: 01/23/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
Gut microbial β-glucuronidase (GUS) plays a key role in metabolizing compounds and influencing disease and drug metabolism, highlighting the need for potent inhibitors to improve drug efficacy and intestinal health. To identify Escherichia coli β-glucuronidase (EcGUS) inhibitors, we designed and synthesized fifty 1,2-benzoselenazol-3-one (BSEA) derivatives using a bioisosterism strategy. Among these, twenty-five BSEA derivatives demonstrated greater inhibitory efficacy than the most potent known EcGUS inhibitor, amoxapine (AMX), with compound 49 showing the strongest activity, achieving an IC50 of 12.9 nM. Structure-inhibitory activity relationship analysis suggested that modifications such as adding benzene rings or nitrogenous heterocycles to the BSEA scaffold enhanced inhibitory activity, influenced by the type and position of substituents. The LC-MS analysis confirmed that compounds 31 and 49 covalently modify Cys197 in EcGUS, and additional covalent linkage of compound 49 was observed on Cys28 and Cys443. In addition, the jump dilution assays proved that compounds 31 was irreversible covalent inhibitors, and its kinetic parameter kinact/KI were determined to be 21292.9 M-1s-1. The compounds 49 was reversible covalent inhibitors and its apparent steady-state inhibition constant Ki∗app were determined to be 23.33 nM. Molecular docking predicted specific interactions, such as hydrogen bonds involving Se and the pyrazole NH of compound 49 with Cys28 and Cys449, which may contribute to its inhibitory action. This study reports the first discovery of covalent inhibitors for EcGUS, with optimized BSEA derivatives acting as novel allosteric covalent inhibitors, revealing structure-activity relationships and molecular determinants that establish their potential in drug development.
Collapse
Affiliation(s)
- Ti-Ti Ying
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hao-Qiang Hu
- Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiao-Wen Wu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xu-Liang Xu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Jian Lv
- Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Shu-Ning Zhang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 201210, China
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China; Binjiang Institute of Artificial Intelligence, ZJUT, Hangzhou, 310051, China
| | - Wei Hou
- Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang Key Laboratory of Green, Low-carbon, and Efficient Development of Marine Fishery Resources, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Guo-Wu Rao
- Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
88
|
Kushawaha AK, Nandi R, Jaiswal AK, Bhatt H, Katiyar S, Ansari A, Choudhury AD, Ali S, Mishra S, Bhatta RS, Srivastava M, Mishra S, Sashidhara KV. Design, synthesis and evaluation of novel carboline-triazole hybrids as promising antimalarial agents. Eur J Med Chem 2025; 290:117514. [PMID: 40120498 DOI: 10.1016/j.ejmech.2025.117514] [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: 12/19/2024] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
A series of twenty-four carboline-triazole derivatives were synthesized using a molecular hybridization approach and evaluated for their antimalarial activity against Plasmodium parasites. Ten compounds exhibited strong in vitro antimalarial activity against both chloroquine-sensitive Pf3D7 and chloroquine-resistant PfK1 strains, with IC50 values ranging from 0.21 to 0.98 μM and 0.32-0.82 μM, respectively. Among them, compound 15k demonstrated significant in vivo antimalarial activity against P. yoelii N67 in Swiss mice. Notably, compounds 15e, 15f, and 15k also inhibited P. berghei liver stage development with IC50 values ranging from 4.81 to 7.58 μM. Evaluation of in vitro pharmacokinetic parameters revealed that the synthesized carboline-triazole conjugate 15k fulfils the criteria for orally active drug development. These findings highlight the potential of this scaffold as a promising framework for the development of antimalarial agents.
Collapse
Affiliation(s)
- Ajay Kishor Kushawaha
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Rohini Nandi
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 Uttar Pradesh, India
| | - Arvind Kumar Jaiswal
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Hemlata Bhatt
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 Uttar Pradesh, India
| | - Sarita Katiyar
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 Uttar Pradesh, India
| | - Alisha Ansari
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 Uttar Pradesh, India
| | - Abhijit Deb Choudhury
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Shabeer Ali
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Shikha Mishra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Rabi Sankar Bhatta
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 Uttar Pradesh, India; Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Mrigank Srivastava
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 Uttar Pradesh, India
| | - Satish Mishra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 Uttar Pradesh, India.
| | - Koneni V Sashidhara
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002 Uttar Pradesh, India; Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031,Uttar Pradesh, India.
| |
Collapse
|
89
|
Saleh NS, El-Sayed NNE, Saleh OA, Allam HA, Mohamed NM, Abbas SES, Said MF. 6,7-Dimethoxy-2-methyl-4-substituted quinazolines: Design, synthesis, EGFR inhibitory activity, in vitro cytotoxicity, and in silico studies. Eur J Med Chem 2025; 290:117502. [PMID: 40120497 DOI: 10.1016/j.ejmech.2025.117502] [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: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Six series of 2,4,6,7-tetrasubstituted quinazolines 4a-c, 6a-c, 8a-c, 10a-d, 13a-d along with quinazoline-tetrahydropyrimidine hybrids 15a-c were designed and synthesized based on keeping the essential key binding pattern of some EGFR inhibitors to appraise their EGFR inhibition and anticancer activity. Twelve compounds out of twenty displayed a significant EGFR inhibition in a subnanomolar level (IC50 = 0.143-0.946 nM) compared to afatinib (IC50 = 0.102 nM). The most potent derivatives 4a, 6c, 8b, 13a and 15b (IC50 = 0.143-0.313 nM) were further screened for their anticancer activity against lung (A549) and colon (HCT116) cancer cell lines, in addition to, normal fibroblast cells (WI-38). It was found that, compounds 6c and 13a show a nearly equipotent to superior cytotoxicity towards (A549) (IC50 = 0.020 and 0.006 μM; respectively) and (IC50 = 0.020 and 0.038 μM; respectively) against HCT116 in comparison to afatinib (IC50 = 0.025 and 0.030 μM; respectively). Also, compounds 6c and 13a caused a cell cycle arrest at S phase and induced apoptosis in A549 and HCT116; respectively. Moreover, in silico studies clarified the binding pattern of the potent compounds in EGFR enzyme active site and confirmed their ability to gratify the structural features meted for binding and rationalized their selectivity. Furthermore, the most active candidates possess promising predicted pharmacokinetic properties.
Collapse
Affiliation(s)
- Nermin S Saleh
- Egyptian Drug Authority (EDA), 51 Wezaret El-Zeraa St., Giza, 35521, Egypt
| | - Nahed N E El-Sayed
- Egyptian Drug Authority (EDA), 51 Wezaret El-Zeraa St., Giza, 35521, Egypt
| | - Ola A Saleh
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical Industries Research Institute, National Research Centre, Egypt
| | - Heba Abdelrasheed Allam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, P.O. Box 11562, Kasr El-Aini Street, Cairo, Egypt
| | - Nada M Mohamed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Modern University for Technology and Information MTI, Cairo, Egypt
| | - Safinaz E-S Abbas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, P.O. Box 11562, Kasr El-Aini Street, Cairo, Egypt
| | - Mona F Said
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, P.O. Box 11562, Kasr El-Aini Street, Cairo, Egypt.
| |
Collapse
|
90
|
Chasák J, Janicki I, Brulíková L. The Liebeskind-Srogl cross-coupling reaction towards the synthesis of biologically active compounds. Eur J Med Chem 2025; 290:117526. [PMID: 40184777 DOI: 10.1016/j.ejmech.2025.117526] [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: 02/19/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/07/2025]
Abstract
In this review, we emphasize the significance of the Liebeskind-Srogl cross-coupling reaction, a palladium-catalyzed process involving the reaction between a thioester and a boronic acid. This reaction has emerged as a fundamental technique in synthetic methodologies aimed at the development of biologically active compounds. The Liebeskind-Srogl cross-coupling method has become an essential approach in chemistry, facilitating the diversification of complex structures that would be significantly more challenging to synthesize through alternative approaches. In this review, we aim to outline the numerous possibilities for preparing a wide range of derivatives, each with notable biological potential.
Collapse
Affiliation(s)
- Jan Chasák
- Department of Organic Chemistry, Faculty of Science, Palacký University, 17. listopadu 12, 77146, Olomouc, Czech Republic
| | - Ignacy Janicki
- Department of Organic Chemistry, Faculty of Science, Palacký University, 17. listopadu 12, 77146, Olomouc, Czech Republic
| | - Lucie Brulíková
- Department of Organic Chemistry, Faculty of Science, Palacký University, 17. listopadu 12, 77146, Olomouc, Czech Republic.
| |
Collapse
|
91
|
Mao Q, Zhang P, Lin F, Fu X, Zhang B, Liu C, Liu Z, Chen X, Dai X, Yue X, Shi X, Pang J, Wang S. Design, synthesis and biological evaluation of 2-[1-(pyridin-2-ylmethyl)-1H-pyrazole-3-carboxamido]benzoic acids as promising urate transporter 1 inhibitors with potential nephroprotective efficacy for the treatment of hyperuricemic nephropathy. Eur J Med Chem 2025; 290:117507. [PMID: 40101451 DOI: 10.1016/j.ejmech.2025.117507] [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: 01/05/2025] [Revised: 03/01/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Abstract
Hyperuricemic nephropathy (HN) is considered an important risk factor for mortality in patients with hyperuricemia. Reducing serum uric acid (UA) levels and mitigating kidney injury are essential components in the treatment of HN. Thus, UA-lowering drugs that can also protect the kidneys are urgently needed. We identified a urate transporter 1 (URAT-1) inhibitor, T29, with cytoprotective efficacy through screening an internal library against hyperuricemia using a UA-induced HK-2 cell injury model. A bioisosteric strategy was then employed to replace the indole core of T29 with pyrazole moieties; this resulted in a series of 2-[1-(pyridin-2-ylmethyl)-1H-pyrazole-3-carboxamido]benzoic acids. Among them, compound 18 demonstrated the best cytoprotective efficacy (cell viability = 92.2 % vs. model = 31.5 %), and the IC50 value of compound 18 against URAT-1 was 3.36 μM; both of these values exceeded T29. In an HN mice model induced by a 0.75 % adenine diet and intraperitoneal injection of potassium oxonate (400 mg/kg), compound 18 significantly reduced the serum UA levels by inhibiting URAT-1 activity. Furthermore, compound 18 improved kidney function by lowering serum creatinine (CRE) and urea nitrogen (BUN) levels while attenuating tubular dilation and inflammatory cell infiltration in the kidneys. Additionally, it suppressed the release of the proinflammatory cytokines IL-1β and TNF-α and reduced kidney fibrosis by downregulating the expression of α-SMA and TGF-β. In conclusion, compound 18 ameliorated HN by inhibiting URAT-1, alleviating immune-inflammatory responses and mitigating fibrosis; the results from this study demonstrate its potential as a therapeutic agent for HN.
Collapse
Affiliation(s)
- Qing Mao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Peng Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Fengwei Lin
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Xuefeng Fu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Bing Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Chang Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Ziyuan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Xing Chen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Xiwen Dai
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China
| | - Xiaoyi Yue
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Xiang Shi
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| | - Jianxin Pang
- Guangdong Provincial Key Laboratory of Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Shaojie Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
92
|
Melfi F, D'Agostino I, Carradori S, Carta F, Angeli A, Costa G, Renzi G, Čikoš A, Vullo D, Rešetar J, Ferraroni M, Baroni C, Mancuso F, Gitto R, Ambrosio FA, Marchese E, Torcasio R, Amodio N, Capasso C, Alcaro S, Supuran CT. O-derivatization of natural tropolone and β-thujaplicin leading to effective inhibitors of human carbonic anhydrases IX and XII. Eur J Med Chem 2025; 290:117552. [PMID: 40179613 DOI: 10.1016/j.ejmech.2025.117552] [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: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Herein we report the chemical derivatization of the naturally occurring Tropolone (TRP) and its related compound β-Thujaplicin (β-TJP) as well as their in vitro assessment for inhibition of the physio/pathologically relevant hCAs isoforms I, II, VA; VII, IX and XII to obtain a first set of inhibition data useful for driving selected derivatives towards appropriate biomedical exploitation. The selected compound 17β was characterized for its chemical stability and assessed for its antiproliferative activity on a multiple myeloma model and showed potent pro-apoptotic features jointly with a safe toxicity profile on healthy cells. The binding mode of β-TJP within the hCA II was assessed by means of X-ray crystallography of the hCA II/β-TJP complex and showed almost complete superposition with the hCA II/TRP adduct reported in the literature. The data produced were used to elaborate a binding prediction model of such compounds on the hCAs VA, IX, and XII which are directly connected to important diseases. Overall, the achievements reported in this work are in the sustainment of the exploitation of naturally occurring troponoloid-based structures for biomedical purposes and thus contribute to the field in extending the variety of available chemical features.
Collapse
Affiliation(s)
- Francesco Melfi
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | | | - Simone Carradori
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy.
| | - Fabrizio Carta
- Neurofarba Department, University of Florence, Sesto Fiorentino, 50019, Florence, Italy.
| | - Andrea Angeli
- Neurofarba Department, University of Florence, Sesto Fiorentino, 50019, Florence, Italy
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università"Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy; Net4Science Academic Spin-Off, Università"Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy
| | - Gioele Renzi
- Neurofarba Department, University of Florence, Sesto Fiorentino, 50019, Florence, Italy
| | - Ana Čikoš
- NMR Centre, Ruđer Bošković Institute, Bijenička cesta 54, 10000, Zagreb, Croatia
| | - Daniela Vullo
- Neurofarba Department, University of Florence, Sesto Fiorentino, 50019, Florence, Italy
| | - Josip Rešetar
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Marta Ferraroni
- 'Ugo Schiff' Chemistry Department, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| | - Chiara Baroni
- 'Ugo Schiff' Chemistry Department, University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Florence, Italy
| | - Francesca Mancuso
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale F. D'Alcontres 13, 98166, Messina, Italy
| | - Rosaria Gitto
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale F. D'Alcontres 13, 98166, Messina, Italy
| | - Francesca Alessandra Ambrosio
- Dipartimento di Scienze della Salute, Università"Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy
| | - Emanuela Marchese
- Dipartimento di Scienze della Salute, Università"Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy
| | - Roberta Torcasio
- Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy; Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, 87036, Rende, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, University "Magna Græcia" of Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy
| | - Clemente Capasso
- Department of Biology, Agriculture and Food Sciences, Institute of Biosciences and Bioresources, CNR, Napoli, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università"Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy; Net4Science Academic Spin-Off, Università"Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100, Catanzaro, Italy
| | - Claudiu T Supuran
- Neurofarba Department, University of Florence, Sesto Fiorentino, 50019, Florence, Italy
| |
Collapse
|
93
|
Ragab A, Ayman R, Salem MA, Ammar YA, Abusaif MS. Unveiling a novel pyrazolopyrimidine scaffold as a dual COX-2/5-LOX inhibitor with immunomodulatory potential: Design, synthesis, target prediction, anti-inflammatory activity, and ADME-T with docking simulation. Eur J Med Chem 2025; 290:117499. [PMID: 40101450 DOI: 10.1016/j.ejmech.2025.117499] [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: 02/13/2025] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/20/2025]
Abstract
Dual-target COX-2/5-LOX inhibitors are regarded as a rational strategy for the design of potent anti-inflammatory agents with favorable safety profiles. In this study, novel pyrazolo[1,5-a]pyrimidine derivatives were synthesized, developed, and screened for their ability to inhibit the cyclooxygenase-2 enzyme in vitro, with comparisons made to the established inhibitors Celecoxib and Meloxicam. Spectroscopic analyses confirmed the structure of the designed derivatives. The target prediction using AI was performed to identify potential targets that could be engaged through Swiss target prediction database. The SAR study was established by incorporating various substituents and nuclei into the pyrazolopyrimidine pharmacophore. The synthesized pyrazolopyrimidines exhibited IC50 values ranging from 53.32 ± 4.43 to 254.90 ± 6.45 nM, in comparison to Celecoxib (IC50 = 6.73 ± 5.69 nM) and Meloxicam (IC50 = 52.35 ± 6.66 nM). Notably, compound 5a was identified as the most active derivative, demonstrating an IC50 of 53.32 ± 4.43 nM. The three most prominent pyrazolopyrimidine derivatives, 3a, 5a, and 6a, were subsequently evaluated for their ability to inhibit the COX-1 and 5-LOX enzymes. Compounds 3a, 5a, and 6a demonstrated inhibitory activity against COX-1, with IC50 values of 476.45 ± 16.56, 757.51 ± 2.61, and 169.13 ± 5.77 nM, respectively. These derivatives 3a, 5a, and 6a showed significant selectivity index values of 7.91, 14.20, and 2.80, respectively, toward COX-2 rather than COX-1 in comparison to Meloxicam (SI = 0.75) and Celecoxib (SI = 2.35). Moreover, compound 5a exhibited 86 % inhibition compared to Zileuton's 88 %, while compounds 3a and 6a displayed inhibition rates of 84 % and 80 %, respectively, at a concentration of 100 μM. The most potent compound 5a, demonstrated the highest 5-LOX inhibitory activity, with IC50 of 2.292 ± 0.14 μM. The most promising pyrazolopyrimidine derivative 5a demonstrated a down-regulation of TNF-α and IL-6 gene expression by approximately 0.3826-fold and 0.2732-fold, respectively, when compared to Celecoxib, which induced reductions of 0.2320-fold and 0.2730-fold in these cytokines to promote apoptosis in RAW264.7 cells. Finally, in-silico ADME-T and docking simulations were conducted to predict the oral bioavailability, toxicity, and binding interactions with binding affinity.
Collapse
Affiliation(s)
- Ahmed Ragab
- Department of Chemistry, Faculty of Science (boys), Al-Azhar University, Nasr City, 11884, Cairo, Egypt; Chemistry Department, Faculty of Science, Galala University, Galala City, 43511, Suez, Egypt.
| | - Radwa Ayman
- Department of Chemistry, Faculty of Science (Girls), Al-Azhar University, Nasr City, Cairo, Egypt.
| | - Mohamed A Salem
- Department of Chemistry, Faculty of Science and Arts, King Khalid University, Mohail, Assir, Saudi Arabia
| | - Yousry A Ammar
- Department of Chemistry, Faculty of Science (boys), Al-Azhar University, Nasr City, 11884, Cairo, Egypt
| | - Moustafa S Abusaif
- Department of Chemistry, Faculty of Science (boys), Al-Azhar University, Nasr City, 11884, Cairo, Egypt.
| |
Collapse
|
94
|
Liu M, Fang T, Wang S, Ma H, Kong L, Deng X, Teng Z, Wang J, Zhang P, Xu L. Repurposing tavaborole to combat resistant bacterial infections through competitive inhibition of KPC-2 and metabolic disruption. Bioorg Chem 2025; 159:108421. [PMID: 40179579 DOI: 10.1016/j.bioorg.2025.108421] [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: 01/12/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
The rise of carbapenem-resistant Enterobacteriaceae (CRE) strains has emerged as an increasing threat to global public health. The development of antibiotic adjuvants presents an economical and promising approach to address this crisis. Through a high-throughput screen of the FDA-approved compound library, we identified tavaborole (AN2690) as a broad-spectrum β-lactamase inhibitor. The mechanistic study revealed that tavaborole formed a reversible binding with the active serine of KPC-2, showing effective competitive inhibition. Its electron-deficient boron atom formed a borate ester bond with hydroxyl group of the serine residue at the active site of KPC-2, transitioning to an sp3-hybridized state that mimicked the tetrahedral intermediate during KPC-2 catalytic. Moreover, transcriptomic analysis and bacterial metabolism assays further unveiled tavaborole addition can inhibit tricarboxylic acid (TCA) cycle, coupled with downregulation of intracellular ATP levels, indicating that tavaborole compromised the bacterial metabolic homeostasis and exerted synergistic antibacterial activity. Notably, the combination treatment further suppressed the development of meropenem resistance. In mouse intraperitoneal infection models, tavaborole effectively restored the efficacy of meropenem against CRE bacteria. These findings elucidate the synergistic mechanisms of tavaborole, expand its potential applications in anti-infection therapeutics, and provide a promising strategy for addressing CRE infections.
Collapse
Affiliation(s)
- Minda Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China; State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Tianqi Fang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; Department of Food Science, College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Shanshan Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hongxia Ma
- College of Animal Science and Technology, Jilin Agricultural University, The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun 130118, China
| | - Lingcong Kong
- College of Animal Science and Technology, Jilin Agricultural University, The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun 130118, China
| | - Xuming Deng
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China; State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zihao Teng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jianfeng Wang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China; State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Peng Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China.
| | - Lei Xu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China; State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
95
|
Gu T, Raval R, Bashkin Z, Zhou C, Ko S, Kong N, Hong S, Bhaskara A, Shah S, Joshi A, Thellakal S, Rim K, Marimuthu A, Venkatesan S, Wang E, Li S, Jayabalan A, Tao A, Fang Y, Xia L, Chui A, Shu E, Zhang T, Chen Z, Njoo E. Synthesis, antiproliferative activity, and biological profiling of C-19 trityl and silyl ether andrographolide analogs in colon cancer and breast cancer cells. Bioorg Med Chem Lett 2025; 121:130163. [PMID: 40043819 DOI: 10.1016/j.bmcl.2025.130163] [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: 10/14/2024] [Revised: 02/08/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Andrographolide, a labdane diterpenoid isolated from Andrographis paniculata, putatively functions through covalent inhibition of NF-κB, a transcription factor that modulates tumor survival and metastasis. Previous studies have found that functionalization of the C-19 hydroxyl alters the primary mode of action from inhibition of NF-κB to the modulation of the Wnt1/β-catenin signaling pathway. Here, we synthesized a series of twelve C-19 trityl and silyl ether analogs, including three novel substituted trityl analogs and four novel substituted silyl analogs of andrographolide. MTT assays revealed cell line selectivity between colorectal and breast cancer cells, which is consistent with known mechanisms of β-catenin-driven cell proliferation in colorectal cancer cell lines. Most compounds exhibited cell line specific antiproliferative activity in HCT-116 and HT-29 colorectal cancer cell lines. Specifically, within 24 h, C-19 analogs of andrographolide exhibit far more limited antiproliferative activity in MCF-7 breast cancer cells compared to HCT-116, HT-29, and MDA-MB-231 cells. Through in vitro TNF-α-dependent NF-κB reporter and Wnt1-dependent luciferase reporter assays, we observed that several analogs generally exhibit greater inhibitory activity compared to andrographolide. Fluorescence imaging demonstrated that cells treated with andrographolide and its C-19 analogs retained similar distributions of active β-catenin, but notable differences in antiproliferative potency upon co-delivery with GSK-3β inhibitor CHIR99021 indicate that several lead compounds exhibit attenuated biological activity selectively in HT-29 cells. Collectively, this work indicates that modest structural modifications at C-19 of andrographolide can have profound implications for its biological activity in mechanisms connected to its anticancer activity.
Collapse
Affiliation(s)
- Tiffany Gu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Rushika Raval
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Zachary Bashkin
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Carina Zhou
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Sanghyuk Ko
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Natalie Kong
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Seoyeon Hong
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Aditya Bhaskara
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Samarth Shah
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Aditi Joshi
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Samahith Thellakal
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Kaitlyn Rim
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Anushree Marimuthu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Srishti Venkatesan
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Emma Wang
- Department of Computer Science & Engineering, Aspiring Scholars Directed Research Program, USA
| | - Sophia Li
- Department of Computer Science & Engineering, Aspiring Scholars Directed Research Program, USA
| | - Aditi Jayabalan
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Alice Tao
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Yilin Fang
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Lorelei Xia
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Aidan Chui
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Emily Shu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Tracy Zhang
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Zhan Chen
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Edward Njoo
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA.
| |
Collapse
|
96
|
Camargo LC, Burgel P, Cardador CM, Mello VC, Rodrigues de Paiva KL, Simões MM, Araújo de Castro RJ, Valente IM, Farias GR, de Castro TB, Muehlmann LA, Báo SN, Longo JPF. Control of aggressive 4T1-luc metastatic breast cancer using immunogenic cell lysates generated with methotrexate. Biomed Pharmacother 2025; 187:118079. [PMID: 40280032 DOI: 10.1016/j.biopha.2025.118079] [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: 01/24/2025] [Revised: 04/01/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
This study investigated a novel immunization therapy for pre-clinical aggressive metastatic breast cancer using immunogenic cell lysates derived from 4T1-luc cells treated with cisplatin and methotrexate, addressing the critical need for improved treatments given the poor prognosis associated with breast cancer metastasis and its significant mortality rate. Methotrexate, a conventional cytotoxic agent, demonstrated a previously unrecognized capacity to induce immunogenic cell lysates, presenting a potential drug repositioning opportunity. In a murine model of stage IV metastatic breast cancer, immunization with these lysates significantly reduced primary tumor growth and lung metastasis, as assessed by bioluminescence imaging. Immunization also modulated immune cell populations, reducing splenomegaly and hepatomegaly, and partially reversing the immunosuppressive phenotype associated with 4T1-luc tumor growth, as evidenced by cytokine profiling (IL-6 and IFN-γ) and flow cytometry analysis of CD4 + and CD8 + T cell subpopulations. Specifically, methotrexate-treated lysates induced a significant shift in CD4 + T cells towards an effector phenotype. These findings highlight the potential of this immunotherapy approach to improve breast cancer treatment outcomes and warrant further investigation.
Collapse
Affiliation(s)
- Luana Cristina Camargo
- Institute of Biological Sciences, University of Brasília, Brasília, Brazil; Department of Basic Psychological Processes, Institute of Psychology, University of Brasília, Brasília, Brazil
| | - Pedro Burgel
- Institute of Biological Sciences, University of Brasília, Brasília, Brazil; Faculty of Medicine, University of Brasília, Brasília, Brazil
| | | | | | | | | | | | | | | | | | | | - Sônia Nair Báo
- Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | | |
Collapse
|
97
|
Nie HJ, Fu YJ, Long S, Wang JY, Zhao WS, Zhai LH, Yang YL, Tan MJ, Hu H, Chen XH. Chemoproteomics reveals proteome-wide covalent and non-covalent targets of withaferin A. Acta Pharmacol Sin 2025; 46:1782-1793. [PMID: 39900821 PMCID: PMC12098870 DOI: 10.1038/s41401-024-01468-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/22/2024] [Indexed: 02/05/2025]
Abstract
Withaferin A (WA), a natural product used in traditional medicine, has recently garnered attention because of its diverse pharmacological effects. However, the direct targets responsible for these effects remain elusive. The discovery of targets is usually serendipitous and research has predominantly concentrated on covalent interactions, overlooking non-covalent targets. The unbiased and proteome-wide mapping of WA-interacting proteins in living cells remains largely unexplored. We have developed a chemical proteomics platform that enabled profiling of the covalent/non-covalent interactome and target occupancy in disease-related cells, which was used to reveal the landscape of the targets of WA in triple-negative breast cancer (TNBC) cells. Analysis of the discovered high-occupancy targets suggested that WA was substantially involved in the RNA metabolism pathway, in addition to other biological processes. Moreover, we biochemically validated a selection of previously unknown high-occupancy targets from various important biological pathways, including the non-covalent target MVK and covalent targets HNRNPF and CKAP4, which all play critical roles in TNBC. Collectively, these findings provided a target map for comprehensive understanding of the anti-TNBC activity of WA, and present WA-targetable proteins as new avenues for pharmacological intervention in TNBC. We anticipate that this platform will be applicable for the unbiased profiling of the targets of WA in various other disease-related cell models, as well as for other bioactive electrophilic natural products in different pathophysiological systems.
Collapse
Affiliation(s)
- Hui-Jun Nie
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ying-Jie Fu
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, 475004, China
| | - Shang Long
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia-Yu Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wen-Si Zhao
- School of Medicine, Tongji University, Shanghai, 200433, China
| | - Lin-Hui Zhai
- School of Medicine, Tongji University, Shanghai, 200433, China
| | - Yin-Long Yang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Min-Jia Tan
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Hu
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Xiao-Hua Chen
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
98
|
Darnowski MG, Lanosky TD, Spada AD, Ma J, Paquette AR, Boddy CN. N-alkyl substituted armeniaspirol analogs show potent antibiotic activity and have low susceptibility to resistance. Bioorg Med Chem Lett 2025; 121:130137. [PMID: 39988012 DOI: 10.1016/j.bmcl.2025.130137] [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: 09/26/2024] [Revised: 01/13/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
The armeniaspirol family of antibiotics have been shown to inhibit the ATP-dependent proteases ClpXP and ClpYQ and to disrupt the electrical membrane potential (ΔΨ) bacterial proton motive force. The synthesis and characterization of first generation armeniaspirol analogs shows the N-alkyl group is amenable to modification. Herein we synthesize eleven second generation N-alkyl analogs and show they display excellent antibiotic potency against multiple MRSA strains and retain the ability to disrupt membrane electrical potential. We also show that it is difficult to generate resistant MRSA mutants to these new compounds, making them appealing leads for new antibiotic development.
Collapse
Affiliation(s)
- Michael G Darnowski
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Taylor D Lanosky
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Antonio D Spada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Jason Ma
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - André R Paquette
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Christopher N Boddy
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada.
| |
Collapse
|
99
|
Hussain MS, Eltaib L, Rana AJ, Maqbool M, Ashique S, Alanazi MN, Khan Y, Agrawal M. Exploiting E3 ligases for lung cancer therapy: The promise of DCAF-PROTACs. Pathol Res Pract 2025; 270:156001. [PMID: 40359818 DOI: 10.1016/j.prp.2025.156001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/06/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
Lung cancer remains the leading cause of cancer-related mortality, underscoring the urgent need for novel therapeutic strategies. One emerging approach in drug development targets oncogenic proteins via the ubiquitin-proteasome system (UPS), specifically through proteolysis-targeting chimeras (PROTACs). Among the various E3 ligase complexes, the CRL4 complex-comprising DDB1 and CUL4-associated factors (DCAFs)-has garnered attention for its roles in cellular homeostasis, DNA repair, and oncogenesis. This review explores the therapeutic potential of DCAF-based PROTACs (DCAF-PROTACs) in lung cancer by focusing on the substrate receptors DCAF13, DCAF15, and DCAF16, which mediate CRL4-dependent ubiquitination. We first discuss the dysregulation of DCAF proteins in lung cancer and then elaborate on their mechanistic role in facilitating target-specific protein degradation via DCAF-E3 ligase complexes. Recent studies show that DCAF-PROTACs selectively degrade oncogenic proteins, addressing treatment resistance and tumor heterogeneity. Notably, DCAF13 promotes lung adenocarcinoma by destabilizing p53, while DCAF15-PROTACs target and degrade RBM39 effectively. Additionally, the development of electrophilic PROTACs targeting DCAF16 presents a promising avenue for degrading nuclear proteins. Despite these advancements, several challenges must be addressed prior to clinical translation, including issues related to drug bioavailability, stability, and emerging resistance mechanisms. This review also explores the potential of combination therapies, particularly with immunotherapy, to enhance tumor specificity and therapeutic efficacy. Ultimately, the deployment of DCAF-PROTACs marks a significant advancement in precision oncology, offering a novel and targeted approach to protein degradation-based cancer treatment.
Collapse
Affiliation(s)
- Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Prem Nagar, Dehradun, Uttarakhand 248007, India.
| | - Lina Eltaib
- Department of Pharmaceutics, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Amita Joshi Rana
- College of Pharmacy, Graphic Era Hill University, Bhimtal, Uttarakhand 263136, India
| | - Mudasir Maqbool
- Department of Pharmacology, Government Medical College Baramulla, Jammu and Kashmir 193103, India
| | - Sumel Ashique
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, West Bengal 711316, India
| | - Mashael N Alanazi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Yumna Khan
- Institute of Biotechnology and Genetic Engineering (Health Division), The University of Agriculture, Peshawar, Khyber Pakhtunkhwa 25000, Pakistan
| | - Mohit Agrawal
- Department of Pharmacology, School of Medical & Allied Sciences, K.R. Mangalam University, Gurugram 122103, India
| |
Collapse
|
100
|
Pandey A, Goswami A, Jithin B, Shukla S. Autophagy: The convergence point of aging and cancer. Biochem Biophys Rep 2025; 42:101986. [PMID: 40224538 PMCID: PMC11986642 DOI: 10.1016/j.bbrep.2025.101986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Autophagy, a dynamic intracellular degradation system, is critical for cellular renovation and maintaining equilibrium. By eliminating damaged components and recycling essential molecules, autophagy safeguards cellular integrity and function. The versatility of the autophagy process across various biological functions enable cells to adapt and maintain homeostasis under unfavourable conditions. Disruptions in autophagy can shift a cell from a healthy state to a disease state or, conversely, support a return to health. This review delves into the multifaceted role of autophagy during aging and age-related diseases such as cancer, highlighting its significance as a unifying target with promising therapeutic implications. Cancer development is a dynamic process characterized by the acquisition of diverse survival capabilities for proliferating at different stages. This progression unfolds over time, with cancer cells exploiting autophagy to overcome encountered stress conditions during tumor development. Notably, there are several common pathways that utilize the autophagy process during aging and cancer development. This highlights the importance of autophagy as a crucial therapeutic target, holding the potential to not only impede the growth of tumor but also enhance the patient's longevity. This review aims to simplify the intricate relationship between cancer and aging, with a particular focus on the role of autophagy.
Collapse
Affiliation(s)
- Anchala Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| | | | | | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|