1
|
Prabha S, Choudhury A, Islam A, Thakur SC, Hassan MI. Understanding of Alzheimer's disease pathophysiology for therapeutic implications of natural products as neuroprotective agents. Ageing Res Rev 2025; 105:102680. [PMID: 39922232 DOI: 10.1016/j.arr.2025.102680] [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: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Alzheimer's disease (AD) is a leading cause of dementia, affecting more than 24.3 million people worldwide in 2024. Sporadic AD (SAD) is more common and occurs in the geriatric population, while familial AD (FAD) is rare and appears before the age of 65 years. Due to progressive cholinergic neuronal loss and modulation in the PKC/MAPK pathway, β-secretase gets upregulated, leading to Aβ aggregation, which further activates tau kinases that form neurofibrillary tangles (NFT). Simultaneously, antioxidant enzymes are also upregulated, increasing oxidative stress (OS) and reactive species by impairing mitochondrial function, leading to DNA damage and cell death. This review discusses the classifications and components of several natural products (NPs) that target these signaling pathways for AD treatment. NPs, including alkaloids, polyphenols, flavonoids, polysaccharides, steroids, fatty acids, tannins, and polypeptides derived from plants, microbes, marine animals, venoms, insects, and mushrooms, are explored in detail. A synergistic combination of plant metabolites, together with prebiotics and probiotics has been shown to decrease Aβ aggregates by increasing the production of bioactive compounds. Toxins derived from venomous organisms have demonstrated effectiveness in modulating signaling pathways and reducing OS. Marine metabolites have also shown neuroprotective and anti-inflammatory properties. The cholera toxin B subunit and an Aβ15 fragment have been combined to create a possible oral AD vaccine, that showed enhancement of cognitive function in mice. Insect tea is also a reliable source of antioxidants. A functional edible mushroom snack bar showed an increment in cognitive markers. Future directions and therapeutic approaches for the treatment of AD can be improved by focusing more on NPs derived from these sources.
Collapse
Affiliation(s)
- Sneh Prabha
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Sonu Chand Thakur
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
2
|
Haque A, Alenezi KM, Rasheed MSMA, Rahman MA, Anwar S, Ahamad S, Gupta D. 4,6-Disubstituted pyrimidine-based microtubule affinity-regulating kinase 4 (MARK4) inhibitors: synthesis, characterization, in-vitro activity and in-silico studies. Front Pharmacol 2025; 15:1517504. [PMID: 39902071 PMCID: PMC11788324 DOI: 10.3389/fphar.2024.1517504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/17/2024] [Indexed: 02/05/2025] Open
Abstract
Introduction Alzheimer's disease (AD) is a neurodegenerative disorder that significantly impacts the cognitive function and memory of a person. Despite the significant research efforts, the ability to completely prevent or effectively treat AD and its related dementias remains limited. Protein kinases are integral to AD pathology and represent promising targets for therapeutic intervention. Methods A series of pyrimidine-based compounds 4-(4-(arylsulfonyl)piperazin-1-yl)-6-(thiophen-3-yl)pyrimidine derivatives (8-14) were synthesized and characterised. ATPase inhibition was carried out against the MARK4 enzyme. Molecular docking and molecular dynamics (MD) simulation at 500 ns was carried out against MARK4 (PDB: 5ES1). The drug-likeness feature and toxicity of the molecules were evaluated using QikProp and other tools. Results Compounds were synthesized following a multi-step approach and characterized using multi-nuclear magnetic resonance (1H/13C-NMR) and mass spectrometry. ATPase inhibition assay of the compounds against MARK4 showed an IC50 value in the micromolar (μM) range. The results of the docking studies were consistent with the in-vitro experiments and identified (9) and (14) as the candidates with the highest affinity towards MARK4. MD simulation further supported these results, showing that the binding of ligands stabilises the target protein. Conclusion Using experimental and theoretical approaches, we demonstrated that the reported class of pyrimidine derivatives are an excellent starting point for developing the next-generation anti-AD drugs.
Collapse
Affiliation(s)
- Ashanul Haque
- Department of Chemistry, College of Science, University of Hail, Ha’il, Saudi Arabia
| | - Khalaf M. Alenezi
- Department of Chemistry, College of Science, University of Hail, Ha’il, Saudi Arabia
| | | | - Md. Ataur Rahman
- Chemistry Program, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Shahzaib Ahamad
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| |
Collapse
|
3
|
Alam M, Gulzar M, Akhtar MS, Rashid S, Zulfareen, Tanuja, Shamsi A, Hassan MI. Epigallocatechin-3-gallate therapeutic potential in human diseases: molecular mechanisms and clinical studies. MOLECULAR BIOMEDICINE 2024; 5:73. [PMID: 39725830 PMCID: PMC11671467 DOI: 10.1186/s43556-024-00240-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/27/2024] [Accepted: 10/29/2024] [Indexed: 12/28/2024] Open
Abstract
Green tea has garnered increasing attention across age groups due to its numerous health benefits, largely attributed to Epigallocatechin 3-gallate (EGCG), its key polyphenol. EGCG exhibits a wide spectrum of biological activities, including antioxidant, anti-inflammatory, antibacterial, anticancer, and neuroprotective properties, as well as benefits for cardiovascular and oral health. This review provides a comprehensive overview of recent findings on the therapeutic potential of EGCG in various human diseases. Neuroprotective effects of EGCG include safeguarding neurons from damage and enhancing cognitive function, primarily through its antioxidant capacity to reduce reactive oxygen species (ROS) generated during physiological stress. Additionally, EGCG modulates key signaling pathways such as JAK/STAT, Delta-Notch, and TNF, all of which play critical roles in neuronal survival, growth, and function. Furthermore, EGCG is involved in regulating apoptosis and cell cycle progression, making it a promising candidate for the treatment of metabolic diseases, including cancer and diabetes. Despite its promising therapeutic potential, further clinical trials are essential to validate the efficacy and safety of EGCG and to optimize its delivery to target tissues. While many reviews have addressed the anticancer properties of EGCG, this review focuses on the molecular mechanisms and signaling pathways by which EGCG used in specific human diseases, particularly cancer, neurodegenerative and metabolic diseases. It serves as a valuable resource for researchers, clinicians, and healthcare professionals, revealing the potential of EGCG in managing neurodegenerative disorders, cancer, and metabolic diseases and highlighting its broader therapeutic values.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Mehak Gulzar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Mohammad Salman Akhtar
- Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, Albaha University, Albaha, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, PO Box 173, 11942, Al-Kharj, Saudi Arabia
| | - Zulfareen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Tanuja
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, P.O. Box 346, Ajman, UAE.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
4
|
Zhan X, Zhou H, Deng C, Hua RX, Pan L, Zhang S, Lu H, He S, Wang Y, Ruan J, Zhou C, He J. Genetic variations in NER pathway gene polymorphisms and Wilms tumor risk: A six-center case-control study in East China. IUBMB Life 2024; 76:1392-1402. [PMID: 39415460 DOI: 10.1002/iub.2919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/17/2024] [Indexed: 10/18/2024]
Abstract
The nucleotide excision repair (NER) system is one of the main ways to protect organisms from DNA damage caused by endogenous and exogenous carcinogens. NER deficiency increases genome mutations, chromosomal aberrations, and cancer viability. However, the genetic association between Wilms tumor and NER pathway gene polymorphisms needs to be further validated. We assessed the associations between 19 NER gene polymorphisms and Wilms tumor susceptibility in 416 cases and 936 controls from East China via the TaqMan method. We found that xeroderma pigmentosum group D (XPD) rs238406 and rs13181 significantly decreased the risk of Wilms tumor [adjusted odds ratio (OR) = 0.59, 95% confidence interval (CI) = 0.46-0.75, p <.0001; adjusted OR = 0.63, 95% CI = 0.44-0.89, p = .009, respectively]. Furthermore, the rs751402 and rs2296147 polymorphisms in the xeroderma pigmentosum group G (XPG) gene were significantly correlated with an increased risk for Wilms tumor (adjusted OR = 1.47, 95% CI = 1.03-2.09, p = .034; adjusted OR = 2.14, 95% CI = 1.29-3.56, p = .003, respectively). Expression quantitative trait loci (eQTL) analysis revealed that these four polymorphisms may affect the expression of genes that are adjacent to XPD and XPG. Our study provides evidence that XPD and XPG gene polymorphisms are associated with Wilms tumor risk. Nonetheless, these findings should be confirmed in a larger sample size.
Collapse
Affiliation(s)
- Xueli Zhan
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Haixia Zhou
- Department of Hematology, The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Changmi Deng
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Rui-Xi Hua
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Lingling Pan
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Shouhua Zhang
- Department of General Surgery, Jiangxi Provincial Children's Hospital, Nanchang, China
| | - Hongting Lu
- Department of Pediatric Surgery, Qingdao Women and Children's Hospital, Qingdao, China
| | - Shaohua He
- Department of Pediatric Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Yizhen Wang
- Department of Pathology, Anhui Provincial Children's Hospital, Hefei, China
| | - Jichen Ruan
- Department of Hematology, The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunlei Zhou
- Department of Pathology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Lian W, Yang X, Duan Q, Li J, Zhao Y, Yu C, He T, Sun T, Zhao Y, Wang W. The Biological Activity of Ganoderma lucidum on Neurodegenerative Diseases: The Interplay between Different Active Compounds and the Pathological Hallmarks. Molecules 2024; 29:2516. [PMID: 38893392 PMCID: PMC11173733 DOI: 10.3390/molecules29112516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Neurodegenerative diseases represent a cluster of conditions characterized by the progressive degeneration of the structure and function of the nervous system. Despite significant advancements in understanding these diseases, therapeutic options remain limited. The medicinal mushroom Ganoderma lucidum has been recognized for its comprehensive array of bioactive compounds with anti-inflammatory and antioxidative effects, which possess potential neuroprotective properties. This literature review collates and examines the existing research on the bioactivity of active compounds and extracts from Ganoderma lucidum in modulating the pathological hallmarks of neurodegenerative diseases. The structural information and preparation processes of specific components, such as individual ganoderic acids and unique fractions of polysaccharides, are presented in detail to facilitate structure-activity relationship research and scale up the investigation of in vivo pharmacology. The mechanisms of these components against neurodegenerative diseases are discussed on multiple levels and elaborately categorized in different patterns. It is clearly presented from the patterns that most polysaccharides of Ganoderma lucidum possess neurotrophic effects, while ganoderic acids preferentially target specific pathogenic proteins as well as regulating autophagy. Further clinical trials are necessary to assess the translational potential of these components in the development of novel multi-target drugs for neurodegenerative diseases.
Collapse
Affiliation(s)
- Wenhui Lian
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
| | - Xu Yang
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
| | - Qidong Duan
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
| | - Jie Li
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
| | - Yuting Zhao
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
| | - Chunhui Yu
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
| | - Tianzhu He
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Tianxia Sun
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
| | - Yu Zhao
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
| | - Weinan Wang
- Jilin Ginseng Academy, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (W.L.); (X.Y.); (Q.D.); (J.L.); (Y.Z.); (C.Y.); (T.H.)
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
6
|
Lei Y, Zhang R, Cai F. Role of MARK2 in the nervous system and cancer. Cancer Gene Ther 2024; 31:497-506. [PMID: 38302729 DOI: 10.1038/s41417-024-00737-z] [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: 11/16/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/03/2024]
Abstract
Microtubule-Affinity Regulating Kinase 2 (MARK2), a member of the serine/threonine protein kinase family, phosphorylates microtubule-associated proteins, playing a crucial role in cancer and neurodegenerative diseases. This kinase regulates multiple signaling pathways, including the WNT, PI3K/AKT/mTOR (PAM), and NF-κB pathways, potentially linking it to cancer and the nervous system. As a crucial regulator of the PI3K/AKT/mTOR pathway, the loss of MARK2 inhibits the growth and metastasis of cancer cells. MARK2 is involved in the excessive phosphorylation of tau, thus influencing neurodegeneration. Therefore, MARK2 emerges as a promising drug target for the treatment of cancer and neurodegenerative diseases. Despite its significance, the development of inhibitors for MARK2 remains limited. In this review, we aim to present detailed information on the structural features of MARK2 and its role in various signaling pathways associated with cancer and neurodegenerative diseases. Additionally, we further characterize the therapeutic potential of MARK2 in neurodegenerative diseases and cancer, and hope to facilitate basic research on MARK2 and the development of inhibitors targeting MARK2.
Collapse
Affiliation(s)
- Yining Lei
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China
| | - Ruyi Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China.
| | - Fei Cai
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437100, China.
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China.
| |
Collapse
|
7
|
Lei Y, Chen Y, Zhang S, Wang W, Zheng M, Zhang R. Qingzhuan dark tea Theabrownin alleviates hippocampal injury in HFD-induced obese mice through the MARK4/NLRP3 pathway. Heliyon 2024; 10:e26923. [PMID: 38455533 PMCID: PMC10918207 DOI: 10.1016/j.heliyon.2024.e26923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/11/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
Background Feeding on a high-fat diet (HFD) results in obesity and chronic inflammation, which may have long-term effects on neuroinflammation and hippocampal injury. Theabrownin, a biologically active compound derived from the microbial fermentation of Qingzhuan dark tea, exhibits anti-inflammatory properties and lipid-lowering effects. Nevertheless, its potential in neuroprotection has yet to be investigated. Consequently, this study aims to investigate the neuroprotective effects of Theabrownin extracted from Qingzhuan dark tea, as well as its potential therapeutic mechanisms. Methods Male C57 mice were subjected to an 8-week HFD to induce obesity, followed by oral administration of Theabrownin from Qingzhuan dark tea. Lipid levels were detected by Elisa kit, hippocampal morphological damage was evaluated by HE and Nissl staining, and the expression levels of GFAP, IBA1, NLRP3, MARK4, and BAX in the hippocampus were detected by immunofluorescence (IF), and protein expression levels of NLRP3, MARK4, PSD95, SYN1, SYP, and Bcl-2 were detected by Western Blot (WB). Results Theabrownin treatment from Qingzhuan dark tea prevents alterations in body weight and lipid levels in HFD-fed mice. Furthermore, Theabrownin decreased hippocampal morphological damage and reduced the activation of astrocytes and microglia in HFD-fed mice. Moreover, Theabrownin decreased the expression of MARK4 and NLRP3 in HFD-fed mice. Besides, Theabrownin elevated the expression of PSD95, SYN1, and SYP in HFD-fed obese mice. Finally, Theabrownin prevented neuronal apoptosis, reduced the expression of BAX, and increased the expression of Bcl-2 in HFD-fed obese mice. Conclusions In summary, our current study presents the first demonstration of the effective protective effect of Theabrownin from Qingzhuan dark tea against HFD-induced hippocampal damage in obese mice. This protection may result from the regulation of the MARK4/NLRP3 signaling pathway, subsequently inhibiting neuroinflammation, synaptic plasticity, and neuronal apoptosis.
Collapse
Affiliation(s)
- Yining Lei
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
| | - Yong Chen
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
| | - Shuo Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
| | - Wei Wang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
| | - Min Zheng
- Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
- Hubei Industrial Technology Research Institute of Intelligent Health, Xianning, Hubei, 437100, China
| | - Ruyi Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
| |
Collapse
|
8
|
Ahmed S, Nadeem M, Hussain I, Fatima S, Anwar S, Rizvi MA, Hassan MI, Tabish M. Preparation of nanoformulation of 5-fluorouracil to improve anticancer efficacy: integrated spectroscopic, docking, and MD simulation approaches. J Biomol Struct Dyn 2023; 42:12523-12536. [PMID: 37850451 DOI: 10.1080/07391102.2023.2270704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/08/2023] [Indexed: 10/19/2023]
Abstract
Nanoformulations (NFs) can be used as a novel drug delivery system to treat all cancer types. One of the major drawbacks of conventional anticancer drugs is that they have poor specificity and higher toxicity towards normal cells. 5-fluorouracil (5-FU) is a well-studied anticancer drug that has a significant role in various cancers, specifically colorectal cancer therapy. This study was performed to determine the functional groups, particle size, surface charge, heterogeneity, and stability of the NF. The NFs of 5-FU were prepared through the ultrasonication technique by increasing the surfactant (Tween-80) concentrations. Among all three NFs, nanoformulated 5-FU (n5-FU) showed the most effective particle size (10.72 nm) with a zeta potential of (-4.57 mV). The cytotoxicity and apoptosis profiles confirmed that n5-FU enhanced the anticancer effect of the pure drug in HCT-116 cells, as evident from MTT assay, fluorescence microscopy, and FACS analysis. In HCT-116 cells, the IC50 values of pure and n5-FU were obtained as 41.3 μM and 18.8 μM, respectively, indicating that n5-FU was more effective against the cancer cell line. The cellular uptake study was performed to check the intake of NF in cancer cells. However, the microtubule-affinity regulating kinase-4 (MARK-4), a cancer-target protein, was purified to study the inhibition and interaction studies. The inhibition assay confirmed the inhibitory potential of 5-FU against MARK-4 protein. the multi-spectroscopic, molecular docking and MD simulation studies were performed to analyse the conformational changes, binding studies, intermolecular interactions, and stability of MARK-4 protein upon binding 5-FU. This demonstrates that NF can enhance the effectiveness of anticancer drugs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shahbaz Ahmed
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, UP, India
| | - Masood Nadeem
- Department of Biosciences, Jamia Milia Islamia, New Delhi, India
| | - Irfan Hussain
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, UP, India
| | - Sana Fatima
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, UP, India
| | - Saleha Anwar
- Center for Interdisciplinary Research in Basic Sciences, Jamia Milia Islamia, New Delhi, India
| | | | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Milia Islamia, New Delhi, India
| | - Mohammad Tabish
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, UP, India
| |
Collapse
|