1
|
Matlyuba Jakhonkulovna S, Bahodirova Kamolovna G, Zokirov M, Umida Tajimuratovna B, Yumashev A, Shichiyakh R, Safarova NI, Nargiza Nusratovna A, Esanmuradova N, Muyassar Karimbaevna T, Lazizakhon A, Ishankulov A. Electrochemical biosensors for early detection of Alzheimer's disease. Clin Chim Acta 2025; 572:120278. [PMID: 40185381 DOI: 10.1016/j.cca.2025.120278] [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: 03/15/2025] [Revised: 03/29/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
In recent years, electrochemical biosensors have shown great promise as innovative tools for the early identification of Alzheimer's disease (AD), a neurodegenerative disorder that severely affects cognitive ability and overall quality of life. This comprehensive review aims to consolidate the latest research on the creation and implementation of electrochemical biosensors designed to detect AD-related biomarkers. We examine cutting-edge approaches to surface modification that enhance the attachment of biorecognition molecules, thus enabling the simultaneous identification of multiple biomarkers. This review emphasizes the crucial role that electrochemical biosensors play in the early diagnosis of Alzheimer's disease, highlighting their potential to revolutionize clinical practices by facilitating timely interventions. In the future, research efforts should concentrate on refining these technologies for widespread clinical adoption, ensuring that they meet the needs of both healthcare professionals and patients.
Collapse
Affiliation(s)
| | - Gulnoz Bahodirova Kamolovna
- Department of Scientific Research, Innovations and Scientific and Pedagogical Personnel Training International School of Finance Technology and Science (ISFT Institute), Uzbekistan
| | | | | | - Alexey Yumashev
- Department of Prosthetic Dentistry, Doctor of Medicine, Professor Sechenov First Moscow State Medical University, Russia
| | - Rustem Shichiyakh
- Department of Management, Candidate of Economic Sciences, Associate Professor. Kuban State Agrarian University named after I.T. Trubilin, Krasnodar, Russia
| | - Nasiba I Safarova
- Department of Otorhinolaryngology, Faculty of Postgraduate Education, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | - Nilufar Esanmuradova
- "Tashkent Institute of Irrigation and Agricultural Mechanization Engineers" National Research University, Tashkent, Uzbekistan; Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Tadjibaeva Muyassar Karimbaevna
- Department of Zoology, Human Morphophysiology and Nutrition (PhD), Nukus State Pedagogical Institute Named After Ajiniyaz, Uzbekistan
| | - Alidjanova Lazizakhon
- International Islamic Academy of Uzbekistan, Senior Lecturer of "UNESCO Chair on Religious Studies and the Comparative Study of World Religions", Kadiri st. 11, Tashkent, Uzbekistan
| | - Alisher Ishankulov
- Samarkand State University named after Sharof Rashidov, Uzbekistan; Kimyo International University in Tashkent, Branch Samarkand, Uzbekistan
| |
Collapse
|
2
|
Singh M, Karthikeyan C, Waiker DK, Tiwari A, Shrivastava SK, Sousa SF, Kiriwan D, Martins FG, Moorthy NSHN. Design, synthesis, and pharmacological evaluation of heteroaryl thiol-linked kojic acid derivatives as a novel class of acetylcholinesterase inhibitors for Alzheimer's disease therapy. 3 Biotech 2025; 15:134. [PMID: 40255452 PMCID: PMC12008096 DOI: 10.1007/s13205-025-04295-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/27/2025] [Indexed: 04/22/2025] Open
Abstract
Natural products have long served as versatile templates for discovering lead molecules against various targets of pharmacological interest. Kojic acid, a fungal metabolite epitomizes this versatility as it elicits broad-spectrum biological properties. Described herein is a series of heteroaryl thiol-linked kojic acid derivatives that demonstrate potent acetylcholinesterase (AChE) inhibition along with anti-amyloid-β (Aβ) aggregation activity and blood brain barrier (BBB) permeability highlighting their potential as a novel class of Anti-Alzheimer's therapeutics. Seventeen kojic acid derivatives, synthesized by incorporating three different heterocyclic thiols, were evaluated for in vitro AChE inhibition employing Ellman's method. The most potent analogs identified from the AChE inhibition studies were further evaluated for binding to the peripheral anionic site (PAS) of AChE using the propidium iodide (PI) displacement assay, anti-amyloid-β (Aβ) aggregation inhibition using the thioflavin T assay, and BBB permeability using the PAMPA-BBB assay. Obtained findings indicated that two compounds MS 21-05 and MS 21-11 bearing a 5-methoxybenzo[d]thiazol-2-yl)thio moiety and 5-phenyl-1,3,4-oxadiazol- 2-yl)thio moiety, respectively, elicited potent AChE inhibition (IC₅₀ < 5 µM), moderate anti-Aβ aggregation effects and good BBB permeability. The molecular docking studies of compound MS 21 - 11 along with its molecular dynamics simulations at peripheral anionic site (PAS) of enzyme AChE provided structural insights into the binding mode of these derivatives. Taken together, the findings of this study establish heteroaryl thiol-linked kojic acid derivatives as a valuable molecular framework for developing anti-Alzheimer's therapeutics that target both cholinergic dysfunction and amyloid-β aggregation. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04295-5.
Collapse
Affiliation(s)
- Madan Singh
- Cancept Therapeutics Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, 484887 India
| | - Chandrabose Karthikeyan
- Cancept Therapeutics Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, 484887 India
| | - Digambar Kumar Waiker
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP 221005 India
| | - Akhilesh Tiwari
- Cancept Therapeutics Laboratory, Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, 484887 India
| | - Sushant K. Shrivastava
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP 221005 India
| | - Sérgio F. Sousa
- UCIBIO—Applied Molecular Biosciences Unit, BioSIM-Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, 4200319 Porto, Portugal
| | - Duangnapa Kiriwan
- UCIBIO—Applied Molecular Biosciences Unit, BioSIM-Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, 4200319 Porto, Portugal
| | - Fábio G. Martins
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculdade de Medicina, Universidade do Porto, 4200319 Porto, Portugal
| | | |
Collapse
|
3
|
Wechakorn K, Payaka A, Masoongnoen J, Wattanalaorsomboon S, Sansenya S. Inhibition potential of n-hexadecanoic and oleic acids from edible insects against α-glucosidase, α-amylase, tyrosinase, and acetylcholinesterase: in vitro and in silico studies. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:3701-3711. [PMID: 39797568 DOI: 10.1002/jsfa.14121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/28/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND Edible insects are used for consumption and traditional medicine due to their rich bioactive compounds. This study examined the bioactive compounds and inhibitory effects of crude extracts from Bombyx mori and Omphisa fuscidentalis on α-glucosidase, α-amylase, acetylcholinesterase (AChE), and tyrosinase. Fatty acids, including n-hexadecanoic acid and oleic acid, were identified in the extracts and evaluated for their inhibitory potential against the enzymes in vitro and in silico. RESULTS The total phenolic content of the edible insect extracts correlated with enzyme inhibitory activity. The quercetin and kaempferol content of B. mori ethyl acetate (EtOAc) extract was also closely related to α-amylase inhibitory activity. The EtOAc and hexane extracts of B. mori showed similar inhibition potential to acarbose and tacrine against α-amylase and AChE, respectively. The hexane extract of O. fuscidentalis exhibited comparable tyrosinase inhibitory activity to kojic acid. n-Hexadecanoic acid and oleic acid were the predominant bioactive compounds in all of the extracts. A kinetic study revealed that n-hexadecanoic acid acted as a mixed-type inhibitor against α-amylase, similar to acarbose, whereas oleic acid showed non-competitive inhibition against AChE, unlike tacrine. Docking studies suggested that these fatty acids bind to the active sites of α-amylase and AChE. CONCLUSION The findings suggest that n-hexadecanoic acid and oleic acid from edible insects could be potential candidates for treating diabetes mellitus and Alzheimer's disease. An animal model might be used for further examination to confirm these findings. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Kanokorn Wechakorn
- Department of Chemistry, Faculty of Science and Technology, Rajamangala University of Technology Thanyaburi, Pathum Thani, Thailand
| | - Apirak Payaka
- School of Science, Walailak University, Nakhon Si Thammarat, Thailand
| | - Jintana Masoongnoen
- Department of Chemistry, Faculty of Science and Technology, Rajamangala University of Technology Thanyaburi, Pathum Thani, Thailand
| | - Sukrit Wattanalaorsomboon
- Department of Chemistry, Faculty of Science and Technology, Rajamangala University of Technology Thanyaburi, Pathum Thani, Thailand
| | - Sompong Sansenya
- Department of Chemistry, Faculty of Science and Technology, Rajamangala University of Technology Thanyaburi, Pathum Thani, Thailand
| |
Collapse
|
4
|
Zueva IV, Saifina LF, Gubaidullina LM, Shulaeva MM, Kharlamova AD, Lenina OA, Belyaev GP, Ziganshina AY, Gao S, Tang W, Semenov VE, Petrov KA. Ionic and Non-Ionic Counterparts Based on Bis(Uracilyl)Alkane Moiety with Highest Selectivity Towards Acetylcholinesterase for Protection Against Organophosphate Poisoning and Treating Alzheimer's Disease. Int J Mol Sci 2025; 26:3759. [PMID: 40332440 PMCID: PMC12027946 DOI: 10.3390/ijms26083759] [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: 03/12/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
A series of bisuracils, in which uracil and 3,6-dimethyluracil moieties were bridged with a polymethylene spacer, and the uracil moiety contained a pentamethylene radical with ionic and non-ionic aminobenzyl groups, were synthesised. These bisuracils have been identified as cholinesterase inhibitors with exceptional selectivity for acetylcholinesterase (AChE) over butyrylcholinesterase (BuChE). These bisuracils, which have been identified as highly effective AChE inhibitors, demonstrated activity at nano- and sub-nanomolar concentrations, with exceptional selectivity for AChE over BuChE. In kinetic studies of lead bisuracils 2b and 3c, both compounds exhibited mixed-type inhibition against AChE and BuChE. Additionally, molecular dynamic simulations demonstrated robust and stable interactions of 2b and 3c with the binding sites of their target. Bisuracil 2b showed significant potential for protection of AChE from irreversible inhibition by paraoxon; the most effective dose of 0.01 mg/kg was shown to reduce mortality in paraoxon-poisoned mice. Bisuracil 3c effectively inhibited brain AChE activity, reversing scopolamine-induced amnesia in mice at a dose of 5 mg/kg, which indicates its potential for cognitive enhancement. These findings position ionic bisuracils as promising prophylactics against organophosphate poisoning and non-ionic bisuracils as viable candidates for Alzheimer's disease therapeutics.
Collapse
Affiliation(s)
- Irina V. Zueva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
| | - Liliya F. Saifina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
| | - Liliya M. Gubaidullina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
| | - Marina M. Shulaeva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
| | - Alexandra D. Kharlamova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
| | - Oksana A. Lenina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
| | - Grigory P. Belyaev
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
| | - Albina Y. Ziganshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
| | - Shan Gao
- School of Pharmacy, Anhui Medical University, Hefei 230032, China;
| | - Wenjian Tang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China;
| | - Vyacheslav E. Semenov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
| | - Konstantin A. Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS Arbuzov str., 8, Kazan 420088, Russia (L.F.S.); (L.M.G.); (M.M.S.); (A.D.K.); (O.A.L.); (G.P.B.); (A.Y.Z.); (K.A.P.)
- Graduate School of Biology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya str, Kazan 420008, Russia
| |
Collapse
|
5
|
Hussain MK, Ahmad M, Khatoon S, Khan MV, Azmi S, Arshad M, Ahamad S, Saquib M. Phytomolecules as Alzheimer's therapeutics: A comprehensive review. Eur J Med Chem 2025; 288:117401. [PMID: 39999743 DOI: 10.1016/j.ejmech.2025.117401] [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: 12/12/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
Alzheimer's disease (AD) is a leading neurodegenerative disorder recognized by progressive cognitive decline and behavioral changes. The pathology of AD is characterized by the accumulation of amyloid-β (Aβ) plaques and the hyperphosphorylation of tau protein, which leads to synaptic loss and subsequent neurodegeneration. Additional contributors to disease progression include metabolic, vascular, and inflammatory factors. Glycogen synthase kinase-3β (GSK-3β) is also implicated, as it plays a crucial role in tau phosphorylation and the progression of neurodegeneration. This review provides a comprehensive analysis of various phytomolecules and their potential to target multiple aspects of AD pathology. We examined natural products from diverse classes, including stilbenes, flavonoids, phenolic acids, alkaloids, coumarins, terpenoids, chromenes, cannabinoids, chalcones, phloroglucinols, and polycyclic polyprenylated acylphloroglucinols (PPAPs). The key mechanisms of action of these phytomolecules include modulating tau protein dynamics to reduce aggregation, inhibiting acetylcholinesterase (AChE) to maintain neurotransmitter levels and enhance cognitive function, and inhibiting β-secretase (BACE1) to decrease Aβ production. Additionally, some phytomolecules were found to influence GSK-3β activity, thereby impacting tau phosphorylation and neurodegeneration. By addressing multiple targets, Aβ production, tau hyperphosphorylation, AChE activity, and GSK-3β, these natural products offer a promising multi-targeted approach to AD therapy. This review highlights their potential to develop effective treatments that not only mitigate core pathological features but also manage the complex, multifactorial aspects of AD progression.
Collapse
Affiliation(s)
- Mohd Kamil Hussain
- Department of Chemistry, Govt Raza P.G. College, M.J.P Rohilkahand University, Rampur, Bareilly, 244901, India.
| | - Moazzam Ahmad
- Defence Research & Development Organization, Selection Centre East, Prayagraj, 211001, India
| | | | - Mohsin Vahid Khan
- Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Sarfuddin Azmi
- Scientific Research Center, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Md Arshad
- Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India.
| | - Mohammad Saquib
- Department of Chemistry, University of Allahabad, Prayagraj (Allahabad), 211002, India; Department of Chemistry, G. R. P. B. Degree College, P. R. S. University, Prayagraj (Allahabad), 211010, UP, India.
| |
Collapse
|
6
|
Attia FM, Kassab RB, Ahmed-Farid OA, Abdel Moneim AE, El-Yamany NA. Zinc Oxide Nanoparticles Attenuated Neurochemical and Histopathological Alterations Associated with Aluminium Chloride Intoxication in Rats. Biol Trace Elem Res 2025; 203:2058-2071. [PMID: 38963645 DOI: 10.1007/s12011-024-04292-4] [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: 01/31/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
The present investigation explored the potential neuroprotective role of zinc oxide nanoparticles (ZnONPs) on aluminum chloride (AlCl3)-mediated Alzheimer's disease (AD)-like symptoms. Rats were distributed into four treatment groups equally: control, ZnONPs (4 mg/kg b.wt.), AlCl3 (100 mg/kg b.wt.), and ZnONPs + AlCl3 groups. Rats were treated for 42 consecutive days. ZnONPs injection into AlCl3-treated rats suppressed the development of oxidative challenge in the cortical and hippocampal tissues, as demonstrated by the decreased neuronal pro-oxidants (malondialdehyde and nitric oxide), and the increased glutathione and catalase levels. Additionally, ZnONPs injection showed anti-inflammatory potency in response to AlCl3 by decreasing levels of tumor necrosis factor-α and interleukin-1β. Moreover, pretreatment with ZnONPs prevented neuronal cell loss by decreasing the level of pro-apoptotic caspase-3 and enhancing the anti-apoptotic B cell lymphoma 2. Furthermore, ZnONPs ameliorated the disturbed acetylcholinesterase activity, monoamines (norepinephrine, dopamine, and serotonin), excitatory (glutamic and aspartic acids), and inhibitory amino acids (GABA and glycine) in response to AlCl3 exposure. These findings indicate that ZnONPs may have the potential as an alternative therapy to minimize or prevent the neurological deficits in AD model by exhibiting antioxidative, anti-inflammation, anti-apoptosis, and neuromodulatory effects.
Collapse
Affiliation(s)
- Fatma M Attia
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Rami B Kassab
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt.
- Biology Department, Faculty of Science and Arts, Almakhwah, Al Baha University, Al Baha, Saudi Arabia.
| | | | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Nabil A El-Yamany
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
7
|
Adnan M, Siddiqui AJ, Bardakci F, Surti M, Badraoui R, Patel M. Mechanistic Insights into the Neuroprotective Potential of Aegle marmelos (L.) Correa Fruits against Aβ-Induced Cell Toxicity in Human Neuroblastoma SH-SY5Y Cells. Pharmaceuticals (Basel) 2025; 18:489. [PMID: 40283926 PMCID: PMC12030591 DOI: 10.3390/ph18040489] [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: 02/27/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Amyloid-β (Aβ) plaque accumulation, oxidative stress, and cholinergic dysfunction are hallmarks of Alzheimer's disease (AD), a neurodegenerative disability that progresses over time, ultimately resulting in the loss of neurons. The side effects and limitations of current synthetic drugs have shifted attention toward natural alternatives. This study investigates the ethanolic extract of Aegle marmelos (L.) Corrêa fruits for their antioxidant, AChE-inhibitory, and anti-amyloidogenic properties, as well as their neuroprotective effects against amyloid beta-peptide (Aβ1-42). Methods: Phytochemical constituents were identified through HR-LCMS analysis and their antioxidant (DPPH, FRAP) and neuroprotective activities (AChE inhibition, ThT binding, MTT assay, ROS reduction, MMP restoration, and AD-related gene expression via qRT-PCR) were assessed using SHSY-5Y neuroblastoma cells. Results: The extract revealed the existence of flavonoids, phenols, and other bioactive substances. In vitro assays demonstrated strong antioxidant and AChE-inhibitory activities, while the ThT binding assay showed protection against amyloid-β aggregation. The extract exhibited no cytotoxicity in SHSY-5Y cells, even at a concentration of 500 μg/mL, whereas Aβ1-42 at 20 μM induced significant cytotoxicity. Co-treatment with Aβ1-42 (10 μM and 20 μM) and the extract improved cell viability (˃50%) and reduced ROS levels. Additionally, the extract restored mitochondrial membrane potential in Aβ1-42 treated cells, highlighting its role in preserving mitochondrial function. Conclusions: These findings suggest that A. marmelos fruits serve as a powerful source of natural antioxidants, AChE inhibitors, and anti-amyloidogenic agents, positioning them as a compelling option for AD treatment.
Collapse
Affiliation(s)
- Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
- King Salman Center for Disability Research, Riyadh 11614, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
| | - Fevzi Bardakci
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
| | - Malvi Surti
- Research and Development Cell (RDC), Parul University, Waghodia, Vadodara 391760, Gujarat, India
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Waghodia, Vadodara 391760, Gujarat, India
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
| | - Mitesh Patel
- Research and Development Cell (RDC), Parul University, Waghodia, Vadodara 391760, Gujarat, India
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Waghodia, Vadodara 391760, Gujarat, India
| |
Collapse
|
8
|
Sidhambaram J, Sakayanathan P, Loganathan C, Iruthayaraj A, Thayumanavan P. Esterified Indole-3-propionic Acid: A Novel Inhibitor against Cholinesterase Identified through Experimental and Computational Approaches. ACS OMEGA 2025; 10:9073-9087. [PMID: 40092751 PMCID: PMC11904713 DOI: 10.1021/acsomega.4c08149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/28/2024] [Accepted: 02/20/2025] [Indexed: 03/19/2025]
Abstract
Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are targeted for designing drugs against cognitive dysfunction. Curcumin (CUR) and indole-3-propionic acid (IPA) are known for their neuroprotective activity. The clinical application of CUR is hindered due to poor absorption and bioavailability. Hence, CUR was conjugated with IPA to form the CUR-IPA diester. CUR-IPA inhibition against electric eel AChE (eAChE), human AChE (hAChE), and hBChE was carried out. In silico and molecular dynamics (MD) analyses of the interaction of CUR-IPA with hAChE and hBChE were done. UV-visible spectroscopy (λmax at 415 and 276 nm), NMR spectrum, and ESI/MS/MS [m/z = 711 (M + H)] confirmed CUR-IPA formation. CUR-IPA showed in vitro antioxidant activity. The IC50 values of eAChE, hAChE, and hBChE enzyme inhibition were 5.66, 59.30, and 60.66 μM, respectively. MD simulation-based analysis such as RMSD, RMSF, free-energy calculation, PCA, FEL, and DCCM confirmed the stable binding of CUR-IPA with hAChE and hBChE. Further QM/MM analysis confirmed the stable interaction of CUR-IPA with hAChE and hBChE. Since CUR-IPA showed in vitro inhibition against AChE and BChE, a further neuroprotective effect in in vivo could be studied.
Collapse
Affiliation(s)
| | | | - Chitra Loganathan
- Department
of Prosthodontics and Implantology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences
(SIMATS), Chennai600 077, India
| | - Ancy Iruthayaraj
- Bioinnov
Solutions LLP, Research and Development Center, Salem, Tamil Nadu 636009, India
| | | |
Collapse
|
9
|
Mashhadi-Sharif N, Soodi M, Mirnajafi-Zadeh J, Shadboorestan A, Ebadi-Ramsheh H, Bakhtiarzadeh F. Ferulago angulata Extract Protects against Beta-amyloid-induced Memory Impairment Through Modulation of Monoaminoxidase Enzymes in the Rat's Hippocampus. Neurochem Res 2025; 50:114. [PMID: 40042687 DOI: 10.1007/s11064-025-04365-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 04/26/2025]
Abstract
Ferulago angulata, a medicinal plant of the Apiaceae family, is known for its antioxidative, anti-inflammatory, anti-apoptotic, and neuroprotective effects. This study investigated the protective potential of F. angulata total extract against memory impairment induced by amyloid β-peptide (Aβ25-35) in rats. Aβ25-35 was bilaterally administered into the CA1 region of the hippocampus in male Wistar rats for four consecutive days. Starting from the first surgery day, rats received oral treatment with F. angulata extract (400 mg/kg) or Rivastigmine (1.5 mg/kg) for 16 consecutive days. Morris water maze tests conducted from days 14 to 17 evaluated the learning and memory performance, followed by biochemical and molecular assays on isolated hippocampi. F. angulata extract significantly reversed Aβ-induced deficits in learning and memory. In the Aβ group, monoamine oxidase A and B enzyme activities and gene expression increased, along with elevated acetylcholinesterase gene expression but decreased enzyme activity compared to the control group. Treatment with F. angulata extract mitigated all these abnormalities. Safety assessments revealed no alterations in blood biochemistry or histology following F. angulata extract treatment. In conclusion, our findings suggest that F. angulata extract possesses neuroprotective properties, and ameliorates Aβ-induced memory deficits by modulating monoamine oxidase enzyme activity and acetylcholinesterase levels without significant adverse effects. Thus, this safe herbal extract shows promise for further exploration in Alzheimer's disease research and potential therapeutic interventions.
Collapse
Affiliation(s)
| | - Maliheh Soodi
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
- Institute for natural products and medicinal plants, Tarbiat Modares University, Tehran, Iran.
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hanieh Ebadi-Ramsheh
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Bakhtiarzadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
10
|
Girgin M, Kantarci-Carsibasi N. Queuine as a potential multi-target drug for alzheimer's disease: insights from protein dynamics. J Biomol Struct Dyn 2025; 43:1847-1868. [PMID: 38095566 DOI: 10.1080/07391102.2023.2293262] [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: 09/28/2023] [Accepted: 12/01/2023] [Indexed: 02/01/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with a complex pathogenesis. One promising approach to treating AD is simultaneously targeting multiple aspects of the disease using a multi-target drug (MTD). In this study, multi-target drug (MTD) potential of the nutraceutical molecule Queuine was explored using molecular docking and molecular dynamics (MD) simulations with five different protein targets engaged in AD: AChE, beta-site amyloid precursor protein cleaving enzyme-1 (BACE-1), N-methyl-D-aspartate receptor (NMDAR), monoamine oxidase A (MAO-A), and Synapsin III. Queuine revealed significant binding affinities, the docking scores being -10.1, -5.97, -5.63, -8.40, and -10.56 kcal/mol for AChE, BACE-1, NMDAR, MAO-A, and Synapsin III, respectively. MD simulations showed that Queuine formed stable complexes and preserved its stability throughout the simulation, the backbone fluctuations remaining within 2.5 Å specifically in the case of the BACE-1. Elastic network model simulations and principal component analysis were carried out to illustrate the dynamics of the protein systems. Significant hinge-bending and twisting-type motions that may be relevant to function were observed around the dimerization interfaces or binding sites. Structural clustering based on PCA analysis and cross-correlation maps demonstrated that Queuine binding altered the protein dynamics more drastically in the case of highly mobile proteins NMDAR and MAO-A. We propose that the neuroprotective effect of Queuine may stem from its prominent inhibitory action on enzymes BACE-1 and AChE. Our results suggest that Queuine may serve as a promising MTD candidate for the treatment of AD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Münteha Girgin
- Department of Chemical Engineering, Uskudar University, Istanbul, Turkey
| | | |
Collapse
|
11
|
Pan N, Liu S, Ge X, Zheng Y. Association of hippocampal atrophy with tau pathology of temporal regions in preclinical Alzheimer's disease. J Alzheimers Dis 2025; 104:191-199. [PMID: 39956951 DOI: 10.1177/13872877251314785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
BackgroundHippocampal atrophy is linked to memory and cognitive deficits, preceding clinical diagnosis of mild cognitive impairment (MCI) by decades. Morphometry changes in the hippocampal formation (HF) and their relationship to tau deposition in non-demented individuals remains unclear.ObjectiveTo investigate morphometry changes in the HF and their association with tau deposition in a non-demented cohort.MethodsEighty-three subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) underwent T1-weighted MRI and Tau-PET scans at baseline and longitudinal follow-up. Participants were divided into amyloid-negative (Aβ-) and amyloid-positive (Aβ+) groups. Hippocampal volume/thickness were measured, and associations with tau deposition in temporal regions were examined using multivariable linear regression.ResultsNo significant association was found between the hippocampal volume/thickness and tau deposition of temporal regions for the Aβ- group. For the Aβ+ group, the hippocampal thickness was significantly associated with tau deposition of entorhinal cortex (ERC) for both hemispheres, and temporal pole, inferior temporal, and middle temporal regions for right hippocampi with the longitudinal follow up scans, while no significant association with the baseline scans. It was interesting that there was strong association between the baseline tau deposition of ERC and temporal pole and the longitudinal follow up thickness of left hippocampi, while the associated regions for the right hemisphere were ERC, temporal pole, and inferior temporal regions.ConclusionsHippocampal atrophy may precede cognitive symptoms, with tau deposition in adjacent temporal regions contributing to hippocampal changes. The right HF appears more vulnerable than the left, indicating hemispheric differences in pathology.
Collapse
Affiliation(s)
- Ningning Pan
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, China
| | - Shujuan Liu
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, China
| | - Xinting Ge
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, China
| | - Yuanjie Zheng
- School of Information Science and Engineering, Shandong Normal University, Jinan, Shandong, China
| |
Collapse
|
12
|
Aboelnasr FG, George MY, Nasr M, Menze ET. Silymarin nanoparticles counteract cognitive impairment induced by doxorubicin and cyclophosphamide in rats; Insights into mitochondrial dysfunction and Nrf-2/HO-1 axis. Eur J Pharmacol 2025; 988:177217. [PMID: 39706469 DOI: 10.1016/j.ejphar.2024.177217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Most cancer patients suffer cognitive impairment following chemotherapy, recognized as "chemobrain". Principally, doxorubicin and cyclophosphamide are frequently utilized conjointly for the treatment of several kinds of tumors. Silymarin was reported to possess anti-inflammatory, antioxidant, and neuroprotective impacts. The recent study shed light on the neuroprotective attributes of silymarin against cognitive dysfunction instigated in rats with doxorubicin/cyclophosphamide combination. Unfortunately, silymarin suffers reduced absorption following oral administration. Silymarin was formulated as a nanoemulsion to be administered intranasally. Male rats were allocated into six groups: control, doxorubicin (2 mg/kg, ip) and cyclophosphamide (50 mg/kg, ip), doxorubicin and cyclophosphamide + silymarin (200 mg/kg, oral), doxorubicin and cyclophosphamide + silymarin nanoemulsion (1 mg/kg, intranasal), silymarin (200 mg/kg, oral), and silymarin nanoemulsion (1 mg/kg, intranasal) groups, and treated for 21 days. The amount of silymarin reaching the brain was found to be enhanced following formulated nanoemulsion administration. Doxorubicin and cyclophosphamide caused behavioral, as well as memory deficits indicated by locomotor activity, y maze, and passive avoidance tests. Also, they induced histological alteration in hippocampi and the prefrontal cortices of rats. Besides, chemotherapy caused cognitive impairment assessed by acetylcholinesterase activity elevation. Additionally, caspase-3 augmentation and of nuclear factor erythroid 2-related factor-2 (Nrf-2) and heme oxygenase-1 (HO-1) pathway disturbance were found following chemotherapy treatment. Silymarin treatment opposed such effects via enhancing memory function, preserving brain architecture, and reducing acetylcholinesterase activity and caspase-3 level. Moreover, silymarin treatment improved mitochondrial biogenesis through activation Nrf-2/HO-1 axis. Collectively, silymarin nanoemulsion, at a 200-fold lower dose, can offer an innovative solution for cancer patients globally.
Collapse
Affiliation(s)
- Fatma G Aboelnasr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt.
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566, Cairo, Egypt
| |
Collapse
|
13
|
Knezovic A, Salkovic-Petrisic M. Cholinergic neurotransmission in the brain of streptozotocin-induced rat model of sporadic Alzheimer's disease: long-term follow up. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02887-2. [PMID: 39891708 DOI: 10.1007/s00702-025-02887-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Rats treated intracerebroventricularly with streptozotocin (STZ-icv) develop pathologic features, which resemble those in Alzheimer's disease and have been proposed as a non-transgenic model for sporadic type of the disease (sAD). We aimed to characterize cholinergic transmission in the rat brain as a function of STZ-icv dose and time after the treatment. Acetylcholinesterase (AChE) activity and expression of muscarinic (M1, M4) and nicotinic (α7) receptors, cholin acetyltransferase (ChAT) and glial fibrillary acidic protein (GFAP) were measured in hippocampus (HPC) and parietotemporal cortex (CTX) of STZ-icv and age-matched control rats one week, and one, three, six and nine months after the icv administration of STZ (0.3, 1 and 3 mg/kg), respectively. Cholinergic and astroglial changes were found most pronounced with a highest STZ dose in time-dependent manner. The cortex and hippocampus exhibited specific alterations in cholinergic transmission following STZ-icv administration, with either similar or distinct patterns depending on the parameter observed: increased AChE activity in HPC and invariable in CTX; increased M4 and ChAT levels in both regions; substantial cortical M1 level increment and moderate hippocampal M1 decrement; and decreased α7 levels in both regions, with subsequent increase observed only in HPC. Alterations in cerebral cholinergic neurotransmission in STZ-icv rat model were mostly following a threephasic time pattern: acute response (Phase I), complete/partial compensation (Phase II), and reappearance/progression of changes (Phase III). Staging structure of cholinergic changes in STZ-icv rat model might be speculated to partly correlate with cholinergic pathology in clinical AD stages.
Collapse
Affiliation(s)
- Ana Knezovic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb, 10 000, Croatia.
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, Zagreb, 10 000, Croatia.
| | - Melita Salkovic-Petrisic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb, 10 000, Croatia
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, Zagreb, 10 000, Croatia
| |
Collapse
|
14
|
Adeyemo SA, Ajao MY, Ogundeyi KJ, Femi-Akinlosotu O, Bakre AG. Ameliorative potential of ethanol extract of Calyptrochilum emarginatum leaves on scopolamine-induced amnesia in male swiss mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118731. [PMID: 39182698 DOI: 10.1016/j.jep.2024.118731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Calyptrochilum emarginatum (Afzel. Ex Sw.) Schltr. (Orchidaceae) is a traditional medicinal plant known for its antimicrobial properties and efficacy in managing convulsive fever and menstrual disorders and addressing conditions such as malaria, tuberculosis, and cough. AIM OF THE STUDY The study aims to examine the memory-enhancing and neuroprotective properties of ethanol extract of Calyptrochilum emarginatum leaves (EECEL) in scopolamine-induced amnesia mice model. MATERIALS AND METHODS Forty-two male mice were divided into six groups (n = 7). Group 1 served as control, administered distilled water (10 mL/kg, p. o), group 2 received scopolamine only (3 mg/kg, i. p.), groups 3 to 6 received pretreatments of EECEL (50, 100, and 200 mg/kg, p. o.) and donepezil (1 mg/kg, p. o.) 30 min before scopolamine (3 mg/kg), for seven days. Following treatments, behavioral (learning and memory) assessments were carried out, while biochemical (acetylcholinesterase activity, oxidative stress markers, inflammatory cytokines markers) and histological evaluations were done after euthanasia. RESULTS Scopolamine significantly impaired spatial, long term and recognition memory. Nevertheless, administration of EECEL (50, 100, and 200 mg/kg orally) enhanced memory function in mice, as observed in the Y maze [F (5, 30) = 20.23, p < 0.0001], Morris water maze [F (10, 90) = 3.105, p = 0.0019; [F (5, 30) = 21.13, p < 0.0001]], and novel object recognition tasks [F (5, 30) = 37.22, p < 0.0001)]. Scopolamine-treated mice exhibited significant dysfunction in the cholinergic system, as evidenced by elevated AChE activity [0.099 ± 0.005 vs. 0.063 ± 0.004 mol/min/g] with an elevation in oxidative stress. On the other hand, administration of EECEL counteracted these consequences by reducing AChE activity, mitigating oxidative damage, reducing pro-inflammatory cytokines, and preventing degeneration of neurons. CONCLUSION The results demonstrated that EECEL effectively mitigates scopolamine-induced memory impairment via an oxido-inflammatory mechanism and modulation of the central cholinergic system.
Collapse
Affiliation(s)
- Stella Afolakemi Adeyemo
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo-State, Nigeria.
| | - Mutiu Yombo Ajao
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo-State, Nigeria.
| | - Kehinde Joshua Ogundeyi
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo-State, Nigeria.
| | - Omowunmi Femi-Akinlosotu
- Developmental Neurobiology and Forensic Anatomy Unit, Department of Anatomy, University of Ibadan, Ibadan, Nigeria.
| | - Adewale Ganiyu Bakre
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo-State, Nigeria.
| |
Collapse
|
15
|
Shaker SE, Fayed DB, Shawky H, Farrag EK. Co-administration of Ceratonia siliqua extract nanoparticles promotes the oral bioavailability and neurotherapeutic efficacy of donepezil in a dementia model. J Pharm Pharmacol 2025; 77:153-169. [PMID: 39046816 DOI: 10.1093/jpp/rgae094] [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: 04/09/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND This study aimed to assess the herb-drug interactions between crude/silver nanoparticle (SNP)-loaded carob extract (Car, NCar, respectively) and donepezil-HCl (DPZ) and their impact on neurotherapeutic outcomes in a dementia model. METHODS Carob pods were subjected to ethanol extraction, and their phytoconstituents were chromatographically analysed. SNP-loaded extract was synthesized and characterized, and dementia-like symptoms were induced in Wistar rats by repeated dosing with 175 mg/kg AlCl3 for 60 days, after which the animals were treated with Car, NCar, DPZ, and combinations of Car/NCar-DPZ for 30 days. The effect of carob formulations on DPZ bioavailability was in-silico profiled and the herb-drug interactions were mathematically assessed as combination indices. RESULTS Different formulations significantly improved cognitive/spatial memory functions, restored dysregulated brain redox and cholinergic functions, and markedly inhibited cholinesterase, as reflected by the reduction/absence of amyloid plaques and neurofibrillary tangles. In silico profiling of the major phytoconstituents revealed their non-P-glycoprotein substrate nature and CYP3A4, 2C19, and 2C9 inhibition, which might have improved the oral bioavailability of DPZ. The combination index calculations revealed strong synergy between DPZ and both carob formulations, with the strongest effect exhibited by the DPZ/NCar combination. CONCLUSION The co-administration of carob extract/SNPs represents a promising approach for enhancing the neurotherapeutic efficacy of DPZ.
Collapse
Affiliation(s)
- Sylvia E Shaker
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki 12622, Cairo, Egypt
| | - Dalia B Fayed
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki 12622, Cairo, Egypt
| | - Heba Shawky
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki 12622, Cairo, Egypt
| | - Ebtehal K Farrag
- Therapeutic Chemistry Department, Pharmaceutical Industries and Drug Research Institute, National Research Centre, Dokki 12622, Cairo, Egypt
| |
Collapse
|
16
|
Vishnumurthy RH, Priya MGR, Tiwari P, Solomon VR. In-silico Studies and Antioxidant and Neuroprotective Assessment of Microencapsulated Celecoxib against Scopolamine-induced Alzheimer's Disease. Curr Pharm Des 2025; 31:320-329. [PMID: 39206485 DOI: 10.2174/0113816128298289240723103828] [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: 12/13/2023] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND OBJECTIVE Alzheimer's disease (AD) is an enervating and chronic progressive neurodegenerative disorder. Celecoxib (CXB) possesses efficacious antioxidants and has neuroprotective, anti-inflammatory, and immunomodulatory properties. However, the poor bioavailability of CXB limits its therapeutic utility. Thus, this study aimed to evaluate the microencapsulated celecoxib MCXB for neuroprotection. METHODOLOGY CXB was screened by molecular docking study using AutoDock (version 5.2), and the following proteins, such as 4EY7, 2HM1, 2Z5X, and 1PBQ were selected for predicting its neuroprotective effect. Scopolamine 20 mg/kg/day for approximately 7 days was administered to albino rats. Pure CXB 100 mg/kg/- day and 200 mg/kg/day, and MCXB 100 mg/kg/day and 200 mg/kg/day were administered, respectively. Further, to assess the oxidative stress, the nitric oxide (NO), superoxide dismutase (SOD), catalase, and lipid peroxidation (LPO) were evaluated using chemical methods. The neurochemical biomarkers like AChE, glutamate, and dopamine were evaluated using the ELISA method. Further, the histopathology of brain cells was carried out to assess the neuro-regeneration and neurodegeneration of the neurons. RESULTS There was a significant binding interaction of CXB (score -6.3, -6.5, -5.1, -9.1) and donepezil (score- 5.5, -7.6, -7.0, and -8.6) with AchE (4EY7), β-secretase (2HM1, monoamine oxidase (2Z5X), and glutamate (1PBQ), respectively. MCXB-treated rats (100 mg/kg/day, 200 mg/kg/day) showed increased SOD levels (p < 0.001), whereas NO, catalase, and LPO levels were significantly (p < 0.001) decreased as compared to scopolamine-treated rats. Further, MCXB-treated rats showed a modulatory effect in the level of dopamine and AchE. However, the glutamate level was significantly (p < 0.001) decreased. CONCLUSION In addition to that, histopathological examination of the hippocampus part showed remarkable improvement in brain cells. So, the findings of the results revealed that MCXB, in a dose-dependent manner, showed a neuroprotective effect against scopolamine-induced AD. This effect may be attributed to the activation of cholinergic pathways.
Collapse
Affiliation(s)
| | - M Gnana Ruba Priya
- College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, Karnataka 560111, India
| | - Prashant Tiwari
- College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, Karnataka 560111, India
| | - Viswas Raja Solomon
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy 502 294, India
| |
Collapse
|
17
|
Zhao X, Hu Q, Wang X, Li C, Chen X, Zhao D, Qiu Y, Xu H, Wang J, Ren L, Zhang N, Li S, Gong P, Hou Y. Dual-target inhibitors based on acetylcholinesterase: Novel agents for Alzheimer's disease. Eur J Med Chem 2024; 279:116810. [PMID: 39243456 DOI: 10.1016/j.ejmech.2024.116810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia among the elderly, accounting for 60 %-70 % of cases. At present, the pathogenesis of this condition remains unclear, but the hydrolysis of acetylcholine (ACh) is thought to play a role. Acetylcholinesterase (AChE) can break down ACh transmission from the presynaptic membrane and stop neurotransmitters' excitatory effect on the postsynaptic membrane, which plays a key role in nerve conduction. Acetylcholinesterase inhibitors (AChEIs) can delay the hydrolysis of acetylcholine (ACh), which represents a key strategy for treating AD. Due to its complex etiology, AD has proven challenging to treat. Various inhibitors and antagonists targeting key enzymes and proteins implicated in the disease's pathogenesis have been explored as potential therapeutic agents. These include Glycogen Synthase Kinase 3β (GSK-3β) inhibitors, β-site APP Cleaving Enzyme (BACE-1) inhibitors, Monoamine Oxidase (MAO) inhibitors, Phosphodiesterase inhibitors (PDEs), N-methyl--aspartic Acid (NMDA) antagonists, Histamine 3 receptor antagonists (H3R), Serotonin receptor subtype 4 (5-HT4R) antagonists, Sigma1 receptor antagonists (S1R) and soluble Epoxide Hydrolase (sEH) inhibitors. The drug development strategy of multi-target-directed ligands (MTDLs) offers unique advantages in the treatment of complex diseases. On the one hand, it can synergistically enhance the therapeutic efficacy of single-target drugs. On the other hand, it can also reduce the side effects. In this review, we discuss the design strategy of dual inhibitors based on acetylcholinesterase and the structure-activity relationship of these drugs.
Collapse
Affiliation(s)
- Xingyi Zhao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Qiaoguan Hu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xiaoqian Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Chunting Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xiao Chen
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Dong Zhao
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Yue Qiu
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, 225321, China
| | - Haoyu Xu
- Yangtze River Pharmaceutical (Group) CO., Ltd. NO.1 South Yangtze River Road, Taizhou City, Jiangsu Province, 225321, China
| | - Jiaqi Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Le Ren
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Na Zhang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Shuang Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Ping Gong
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Yunlei Hou
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 105 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
18
|
Aydin BO, Anil DA, Demir Y, Alagoz MA. Organohalogen chalcones: design, synthesis, ADMET prediction, molecular dynamics study and inhibition effect on acetylcholinesterase and carbonic anhydrase. Mol Divers 2024; 28:3739-3755. [PMID: 38170420 DOI: 10.1007/s11030-023-10774-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 11/17/2023] [Indexed: 01/05/2024]
Abstract
In an effort to discover potential acetylcholinesterase (AChE) and carbonic anhydrase (CA) inhibitors, a novel series of organohalogen chalcone derivatives (12-20, 23-30) was synthesized, and their chemical structures were characterized by spectral analysis. They showed a highly potent inhibition effect on AChE and hCAs (Ki values range from 5.07 ± 0.062 to 65.53 ± 4.36 nM for AChE, 13.54 ± 2.55 to 94.11 ± 10.39 nM for hCA I, and 5.21 ± 0.54 to 57.44 ± 3.12 nM for hCA II). In addition, the chalcone derivatives with the highest inhibitor score docked into the active site of the indicated metabolic enzyme receptors, and their absorption, metabolism, and toxic properties were evaluated according to ADMET's estimation.Compounds 16 and 19 exhibited the highest inhibition score, emerged as lead compounds, and inspired the development of more potent compounds.
Collapse
Affiliation(s)
- Busra Ozturk Aydin
- Department of Chemistry, Faculty of Science, Ataturk University, 25240, Erzurum, Turkey
| | - Derya Aktas Anil
- Department of Chemistry, Faculty of Science, Ataturk University, 25240, Erzurum, Turkey.
- Department of Chemistry and Chemical Process Technologies, Technical Sciences Vocational School, Ataturk University, 25240, Erzurum, Turkey.
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational School, Ardahan University, 75002, Ardahan, Turkey
| | - Mehmet Abdullah Alagoz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Inonu University, Malatya, Turkey
| |
Collapse
|
19
|
Ferjančič Benetik S, Knez D, Obreza A, Košak U, Gobec S. Dual inhibition of butyrylcholinesterase and p38α mitogen-activated protein kinase: A new approach for the treatment of Alzheimer's disease. Pharmacol Ther 2024; 264:108748. [PMID: 39521443 DOI: 10.1016/j.pharmthera.2024.108748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
The simultaneous targeting of neuroinflammation and cholinergic hypofunction, the key pathological changes in Alzheimer's disease (AD), is not addressed by drugs currently in clinical trials, highlighting a critical therapeutic gap. We propose that dual-acting small molecules that inhibit butyrylcholinesterase (BChE) and mitogen-activated protein kinase p38α (p38α MAPK) represent a novel strategy to combat AD. This hypothesis is supported by cellular and animal studies as well as in silico modelling showing that it is possible to act simultaneously on both enzymes. Amyloid beta (Aβ) plaques trigger a pro-inflammatory microglial response that overactivates p38α MAPK, leading to increased Aβ synthesis, tau hyperphosphorylation, and altered synaptic plasticity. Overactivated microglia exacerbate neuroinflammation and cholinergic degeneration, ultimately leading to cognitive impairment. Structural similarities between the binding sites of BChE and p38α MAPK provide a promising basis for the development of dual inhibitors that could alleviate AD symptoms and address the underlying pathology.
Collapse
Affiliation(s)
| | - Damijan Knez
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Aleš Obreza
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Urban Košak
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
20
|
Taheri M, Roghani M, Sedaghat R. Metformin Mitigates Trimethyltin-Induced Cognition Impairment and Hippocampal Neurodegeneration. Cell Mol Neurobiol 2024; 44:70. [PMID: 39441380 PMCID: PMC11499442 DOI: 10.1007/s10571-024-01502-4] [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: 04/10/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024]
Abstract
The neurotoxicant trimethyltin (TMT) triggers cognitive impairment and hippocampal neurodegeneration. TMT is a useful research tool for the study of Alzheimer's disease (AD) pathogenesis and treatment. Although the antidiabetic agent metformin has shown promising neuroprotective effects, however, its precise modes of action in neurodegenerative disorders need to be further elucidated. In this study, we investigated whether metformin can mitigate TMT cognition impairment and hippocampal neurodegeneration. To induce an AD-like phenotype, TMT was injected i.p. (8 mg/kg) and metformin was administered daily p.o. for 3 weeks at 200 mg/kg. Our results showed that metformin administration to the TMT group mitigated learning and memory impairment in Barnes maze, novel object recognition (NOR) task, and Y maze, attenuated hippocampal oxidative, inflammatory, and cell death/pyroptotic factors, and also reversed neurodegeneration-related proteins such as presenilin 1 and p-Tau. Hippocampal level of AMP-activated protein kinase (AMPK) as a key regulator of energy homeostasis was also improved following metformin treatment. Additionally, metformin reduced hippocampal acetylcholinesterase (AChE) activity, glial fibrillary acidic protein (GFAP)-positive reactivity, and prevented the loss of CA1 pyramidal neurons. This study showed that metformin mitigated TMT-induced neurodegeneration and this may pave the way to develop new therapeutics to combat against cognitive deficits under neurotoxic conditions.
Collapse
Affiliation(s)
- Mahdieh Taheri
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| | - Reza Sedaghat
- Department of Pathology, School of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
21
|
Wang L, Liu J, Gui W, Zhang R, Li X, Fang L, Li H, Pan D, Ye W. Molecular interaction mechanisms on (-)-epigallocatechin-3-gallate improving activities of inhibited acetylcholinesterase by selected organophosphorus pesticides in vitro & vivo. Sci Rep 2024; 14:22296. [PMID: 39333189 PMCID: PMC11436701 DOI: 10.1038/s41598-024-72637-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 09/09/2024] [Indexed: 09/29/2024] Open
Abstract
(-)-Epigallocatechin-3-gallate (EGCG) is reported to have benefits for the treatment of Alzheimer's disease by binding with acetylcholinesterase (AChE) to enhance the cholinergic neurotransmission. Organophosphorus pesticides (OPs) inhibited AChE and damaged the nervous system. This study investigated the combined effects of EGCG and OPs on AChE activities in vitro & vivo. The results indicated that EGCG significantly reversed the inhibition of AChE caused by OPs. In vitro, EGCG reactived AChE in three group tubes incubated for 110 min, and in vivo, it increased the relative activities of AChE from less than 20% to over 70% in brain and vertebral of zebrafish during the exposure of 34 h. The study also proposed the molecular interaction mechanisms through the reactive kinetics and computational analyses of density functional theory, molecular docking, and dynamic modeling. These analyses suggested that EGCG occupied the key residues, preventing OPs from binding to the catalytic center of AChE, and interfering with the initial affinity of OPs to the central active site. Hydrogen bonding, conjugation, and steric interactions were identified as playing important roles in the molecular interactions. The work suggests that EGCG antagonized the inhibitions of OPs on AChE activities and potentially offered the neuroprotection against the induced damage.
Collapse
Affiliation(s)
- Lijun Wang
- School of Resources and Environment, Anhui Agricultural University, No. 130 West Changjiang Road, Hefei, 230036, China
| | - Jian Liu
- School of Resources and Environment, Anhui Agricultural University, No. 130 West Changjiang Road, Hefei, 230036, China
| | - Wenqian Gui
- School of Resources and Environment, Anhui Agricultural University, No. 130 West Changjiang Road, Hefei, 230036, China
| | - Rong Zhang
- School of Resources and Environment, Anhui Agricultural University, No. 130 West Changjiang Road, Hefei, 230036, China.
| | - Xinmei Li
- School of Resources and Environment, Anhui Agricultural University, No. 130 West Changjiang Road, Hefei, 230036, China
| | - Liancheng Fang
- School of Resources and Environment, Anhui Agricultural University, No. 130 West Changjiang Road, Hefei, 230036, China
| | - Hui Li
- School of Resources and Environment, Anhui Agricultural University, No. 130 West Changjiang Road, Hefei, 230036, China
| | - Dandan Pan
- School of Resources and Environment, Anhui Agricultural University, No. 130 West Changjiang Road, Hefei, 230036, China
| | - Wenling Ye
- School of Resources and Environment, Anhui Agricultural University, No. 130 West Changjiang Road, Hefei, 230036, China
| |
Collapse
|
22
|
Ojo OA, Ajayi-Odoko OA, Gyebi GA, Ayokunle DI, Ogunlakin AD, Ezenabor EH, Olanrewaju AA, Agbeye OD, Ogunwale ET, Rotimi DE, Fouad D, Batiha GES, Adeyemi OS. Network Pharmacology, Molecular Dynamics and In Vitro Assessments of Indigenous Herbal Formulations for Alzheimer's Therapy. Life (Basel) 2024; 14:1222. [PMID: 39459522 PMCID: PMC11508826 DOI: 10.3390/life14101222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative condition marked by amyloid plaques, synaptic dysfunction, and neuronal loss. Besides conventional medical care, herbal therapies, both raw and refined, have attracted researchers for their potential therapeutic effects. As a proof-of-concept, our study combined HPLC-DAD analysis of bioactive constituents, network pharmacology, molecular dynamics (MD), molecular docking, post-MD analysis, and experimental verification to investigate the mechanisms of crude drug formulations as a therapeutic strategy for AD. We identified nine bioactive compounds targeting 188 proteins and 1171 AD-associated genes. Using a Venn diagram, we found 47 overlapping targets, forming "herb-compound-target (HCT)" interaction networks and a protein‒protein interaction (PPI) network. Simulations analyzed binding interactions among the three core targets and their compounds. MD assessed the stability of the best-ranked poses and beneficial compounds for each protein. Among the top 22 hub genes, AChE, BChE, and MAO, ranked 10, 14, and 34, respectively, were selected for further analysis. Two tetraherbal formulations, Form A and Form B, showed notable activity against AChE. Form A exhibited significant (p < 0.0001) inhibitory activity (IC50 = 114.842 ± 2.084 µg/mL) compared to Form B (IC50 = 142.829 ± 4.258 µg/mL), though weaker than galantamine (IC50 = 27.950 ± 0.122 µg/mL). Form B had significant inhibitory effects on BChE (IC50 = 655.860 ± 32.812 µg/mL) compared to Form A (IC50 = 679.718 ± 20.656 µg/mL), but lower than galantamine (IC50 = 23.126 ± 0.683 µg/mL). Both forms protected against Fe2+-mediated brain injury by inhibiting MAO. Docking identified quercetin (-10.2 kcal/mol) and myricetin (-10.1 kcal/mol) for AChE; rutin (-10.6 kcal/mol) and quercetin (-9.7 kcal/mol) for BChE; and kaempferol (-9.1 kcal/mol) and quercetin (-8.9 kcal/mol) for MAO. These compounds were thermodynamically stable based on MD analysis. Collectively, the results offer a scientific rationale for the use of these specifically selected medicinal herbs as AD medications.
Collapse
Affiliation(s)
- Oluwafemi Adeleke Ojo
- Good Health and Wellbeing Research Clusters (SDG 03), Bowen University, Iwo 232102, Nigeria; (A.D.O.); (E.H.E.); (O.D.A.); (O.S.A.)
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Group, Biochemistry Programme, Bowen University, Iwo 232102, Nigeria
| | | | - Gideon Ampoma Gyebi
- Natural Products and Structural (Bio-Chem)-Informatics Research Laboratory (NpsBC-RI), Department of Biochemistry, Bingham University, Karu 961105, Nigeria;
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P.O. Box 1334, Durban 4000, South Africa
| | | | - Akingbolabo Daniel Ogunlakin
- Good Health and Wellbeing Research Clusters (SDG 03), Bowen University, Iwo 232102, Nigeria; (A.D.O.); (E.H.E.); (O.D.A.); (O.S.A.)
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Group, Biochemistry Programme, Bowen University, Iwo 232102, Nigeria
| | - Emmanuel Henry Ezenabor
- Good Health and Wellbeing Research Clusters (SDG 03), Bowen University, Iwo 232102, Nigeria; (A.D.O.); (E.H.E.); (O.D.A.); (O.S.A.)
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Group, Biochemistry Programme, Bowen University, Iwo 232102, Nigeria
| | | | - Oluwatobi Deborah Agbeye
- Good Health and Wellbeing Research Clusters (SDG 03), Bowen University, Iwo 232102, Nigeria; (A.D.O.); (E.H.E.); (O.D.A.); (O.S.A.)
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Group, Biochemistry Programme, Bowen University, Iwo 232102, Nigeria
| | | | - Damilare Emmanuel Rotimi
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA;
- Department of Biochemistry, Landmark University, Omu-Aran 251101, Nigeria
| | - Dalia Fouad
- Department of Zoology, College of Science, King Saud University, P.O. Box 22452, Riyadh 11495, Saudi Arabia;
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutic, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt;
| | - Oluyomi Stephen Adeyemi
- Good Health and Wellbeing Research Clusters (SDG 03), Bowen University, Iwo 232102, Nigeria; (A.D.O.); (E.H.E.); (O.D.A.); (O.S.A.)
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Group, Biochemistry Programme, Bowen University, Iwo 232102, Nigeria
- Laboratory of Sustainable Animal Environment Systems, Graduate School of Agricultural Sciences, Tohoku University, Sendai 980-8579, Japan
| |
Collapse
|
23
|
de la Monte SM, Tong M. Dysregulated mTOR networks in experimental sporadic Alzheimer's disease. Front Cell Neurosci 2024; 18:1432359. [PMID: 39386180 PMCID: PMC11461251 DOI: 10.3389/fncel.2024.1432359] [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: 05/13/2024] [Accepted: 08/29/2024] [Indexed: 10/12/2024] Open
Abstract
Background Beyond the signature amyloid-beta plaques and neurofibrillary tangles, Alzheimer's disease (AD) has been shown to exhibit dysregulated metabolic signaling through insulin and insulin-like growth factor (IGF) networks that crosstalk with the mechanistic target of rapamycin (mTOR). Its broad impact on brain structure and function suggests that mTOR is likely an important therapeutic target for AD. Objective This study characterizes temporal lobe (TL) mTOR signaling abnormalities in a rat model of sporadic AD neurodegeneration. Methods Long Evans rats were given intracerebroventricular injections of streptozotocin (ic-STZ) or saline (control), and 4 weeks later, they were administered neurobehavioral tests followed by terminal harvesting of the TLs for histopathological study and measurement of AD biomarkers, neuroinflammatory/oxidative stress markers, and total and phosphorylated insulin/IGF-1-Akt-mTOR pathway signaling molecules. Results Rats treated with ic-STZ exhibited significantly impaired performance on Rotarod (RR) and Morris Water Maze (MWM) tests, brain atrophy, TL and hippocampal neuronal and white matter degeneration, and elevated TL pTau, AβPP, Aβ, AChE, 4-HNE, and GAPDH and reduced ubiquitin, IL-2, IL-6, and IFN-γ immunoreactivities. In addition, ic-STZ reduced TL pY1135/1136-IGF-1R, Akt, PTEN, pS380-PTEN, pS2448-mTOR, p70S6K, pT412-p70S6K, p/T-pT412-p70S6K, p/T-Rictor, and p/T-Raptor. Conclusion Experimental ic-STZ-induced sporadic AD-type neurodegeneration with neurobehavioral dysfunctions associated with inhibition of mTOR signaling networks linked to energy metabolism, plasticity, and white matter integrity.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Medicine, Pathology and Laboratory Medicine, Neurology, and Neurosurgery, Rhode Island Hospital, Women and Infants Hospital, The Alpert Medical School at Brown University, Providence, RI, United States
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, The Alpert Medical School at Brown University, Providence, RI, United States
| |
Collapse
|
24
|
Yang Y, Tong M, de la Monte SM. Early-Stage Moderate Alcohol Feeding Dysregulates Insulin-Related Metabolic Hormone Expression in the Brain: Potential Links to Neurodegeneration Including Alzheimer's Disease. J Alzheimers Dis Rep 2024; 8:1211-1228. [PMID: 39247872 PMCID: PMC11380283 DOI: 10.3233/adr-240026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
Background Alzheimer's disease (AD), one of the most prevalent causes of dementia, is mainly sporadic in occurrence but driven by aging and other cofactors. Studies suggest that excessive alcohol consumption may increase AD risk. Objective Our study examined the degree to which short-term moderate ethanol exposure leads to molecular pathological changes of AD-type neurodegeneration. Methods Long Evans male and female rats were fed for 2 weeks with isocaloric liquid diets containing 24% or 0% caloric ethanol (n = 8/group). The frontal lobes were used to measure immunoreactivity to AD biomarkers, insulin-related endocrine metabolic molecules, and proinflammatory cytokines/chemokines by duplex or multiplex enzyme-linked immunosorbent assays (ELISAs). Results Ethanol significantly increased frontal lobe levels of phospho-tau, but reduced Aβ, ghrelin, glucagon, leptin, PAI, IL-2, and IFN-γ. Conclusions Short-term effects of chronic ethanol feeding produced neuroendocrine molecular pathologic changes reflective of metabolic dysregulation, together with abnormalities that likely contribute to impairments in neuroplasticity. The findings suggest that chronic alcohol consumption rapidly establishes a platform for impairments in energy metabolism that occur in both the early stages of AD and alcohol-related brain degeneration.
Collapse
Affiliation(s)
- Yiwen Yang
- Molecular Pharmacology, Physiology and Biotechnology Graduate Program, Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Suzanne M. de la Monte
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, the Providence VA Medical Center, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Departments of Neurology and Neurosurgery, Rhode Island Hospital, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
25
|
Khamies SM, El-Yamany MF, Ibrahim SM. Canagliflozin Mitigated Cognitive Impairment in Streptozotocin-Induced Sporadic Alzheimer's Disease in Mice: Role of AMPK/SIRT-1 Signaling Pathway in Modulating Neuroinflammation. J Neuroimmune Pharmacol 2024; 19:39. [PMID: 39073453 DOI: 10.1007/s11481-024-10140-y] [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: 02/12/2024] [Accepted: 07/14/2024] [Indexed: 07/30/2024]
Abstract
Sporadic Alzheimer's disease (SAD) represents a major health concern especially among elderly. Noteworthy, neuroinflammation and oxidative stress are highly implicated in AD pathogenesis resulting in enhanced disease progression. Moreover, most of the available anti-Alzheimer drugs have several adverse effects with variable efficacy, therefore new strategies, including agents with anti-inflammatory and antioxidant effects, are encouraged. Along these lines, canagliflozin (CAN), with its anti-inflammatory and anti-apoptotic activities, presents a promising candidate for AD treatment. Therefore, this study aimed to evaluate the therapeutic potential of CAN via regulation of AMPK/SIRT-1/BDNF/GSK-3β signaling pathway in SAD. SAD model was induced by intracerebroventricular streptozotocin injection (ICV-STZ;3 mg/kg, once), while CAN was administered (10 mg/kg/day, orally) to STZ-treated mice for 21 days. Behavioral tests, novel object recognition (NOR), Y-Maze, and Morris Water Maze (MWM) tests, histopathological examination, total adenosine monophosphate-activated protein kinase (T-AMPK) expression, p-AMPK, and silent information regulator-1 (SIRT-1) were evaluated. Furthermore, brain-derived neurotrophic factor (BDNF), glycogen synthase kinase-3β (GSK-3β), acetylcholinesterase (AChE), Tau protein, insulin-degrading enzyme (IDE), nuclear factor erythroid-2 (Nrf-2), interleukin-6 (IL-6), nuclear factor kappa-B-p65 (NFκB-p65), beta-site APP cleaving enzyme 1 (BACE-1), and amyloid beta (Aβ) plaque were assessed. CAN restored STZ-induced cognitive deficits, confirmed by improved behavioral tests and histopathological examination. Besides, CAN halted STZ-induced neurotoxicity through activation of p-AMPK/SIRT-1/BDNF pathway, subsequently reduction of GSK-3β, Tau protein, AChE, NFκB-p65, IL-6, BACE-1, and Aβ plaque associated with increased IDE and Nrf-2. Consequentially, our findings assumed that CAN, via targeting p-AMPK/SIRT-1 pathway, combated neuroinflammation and oxidative stress in STZ-induced AD. Thus, this study highlighted the promising effect of CAN for treating AD.
Collapse
Affiliation(s)
- Sara M Khamies
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Menoufia University, Menoufia, 32511, Egypt
| | - Mohammed F El-Yamany
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Cairo University, ElKasr Elaini Street, Cairo, 11562, Egypt
| | - Sherehan M Ibrahim
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Cairo University, ElKasr Elaini Street, Cairo, 11562, Egypt.
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Modern University for Technology and Information, Cairo, Egypt.
| |
Collapse
|
26
|
Pereira ME, Lima LS, Souza JV, de Souza da Costa N, da Silva JF, Guiloski IC, Irioda AC, Oliveira CS. Evaluation of the Neuroprotective Effect of Organic Selenium Compounds: An in Vitro Model of Alzheimer's Disease. Biol Trace Elem Res 2024; 202:2954-2965. [PMID: 37803188 DOI: 10.1007/s12011-023-03893-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023]
Abstract
Selenium (Se) is an essential trace element for human health and plays an important role in the development and maintenance of central nervous system functions. Se deficiency has been associated with cognitive decline and increased oxidative stress. The increase in oxidative stress is one of the hypotheses for the emergence and worsening of neurodegenerative diseases, such as Alzheimer's disease (AD). To investigate the neuroprotective effects of organic Se compounds in human neuroblastoma cells (SH-SY5Y) differentiated into cholinergic neurons-like. The SH-SY5Y cells were differentiated into cholinergic neuron-like with retinoic acid (RA) and brain-derived neurotrophic factor (BDNF). AD was mimicked exposing the cells to okadaic acid (OA) and beta-amyloid protein (Aβ). The neuroprotective effect of organic Se compounds, selenomethionine (SeMet) and Ebselen, was evaluated through cell viability tests, acetylcholinesterase and antioxidant enzyme activities, and detection of reactive oxygen species (ROS). None of the SeMet concentrations tested protected against the toxic effect of OA + Aβ. On the other hand, previous exposure to 0.1 and 1 µM Ebselen protected cells from the toxic effect of OA + Aβ. Cell differentiation induced by RA and BDNF exposure was effective, showing characteristics of neuronal cells, and pointing to a promising model of AD. Ebselen showed a protective effect, but more studies are needed to identify the mechanism of action.
Collapse
Affiliation(s)
- Meire Ellen Pereira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Luiza Siqueira Lima
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Júlia Vicentin Souza
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Nayara de Souza da Costa
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Juliana Ferreira da Silva
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Izonete Cristina Guiloski
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | | | - Cláudia Sirlene Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil.
- Faculdades Pequeno Príncipe, Curitiba, PR, Brazil.
| |
Collapse
|
27
|
Garcia Ratés S, García‐Ayllón M, Falgàs N, Brangman SA, Esiri MM, Coen CW, Greenfield SA. Evidence for a novel neuronal mechanism driving Alzheimer's disease, upstream of amyloid. Alzheimers Dement 2024; 20:5027-5034. [PMID: 38780014 PMCID: PMC11247685 DOI: 10.1002/alz.13869] [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: 01/29/2024] [Revised: 03/21/2024] [Accepted: 04/08/2024] [Indexed: 05/25/2024]
Abstract
This perspective offers an alternative to the amyloid hypothesis in the etiology of Alzheimer's disease (AD). We review evidence for a novel signaling mechanism based on a little-known peptide, T14. T14 could drive neurodegeneration as an aberrantly activated process of plasticity selective to interconnecting subcortical nuclei, the isodendritic core, where cell loss starts at the pre-symptomatic stages of the disease. Each of these cell groups has the capacity to form T14, which can stimulate production of p-Tau and β-amyloid, suggestive of an upstream driver of neurodegeneration. Moreover, results in an animal AD model show that antagonism of T14 with a cyclated variant, NBP14, prevents formation of β-amyloid, and restores cognitive function to that of wild-type counterparts. Any diagnostic and/or therapeutic strategy based on T14-NBP14 awaits validation in clinical trials. However, an understanding of this novel signaling system could bring much-needed fresh insights into the progression of cell loss underlying AD. HIGHLIGHTS: The possible primary mechanism of neurodegeneration upstream of amyloid. Primary involvement of selectively vulnerable subcortical nuclei, isodendritic core. Bioactive peptide T14 trophic in development but toxic in context of mature brain. Potential for early-stage biomarker to detect Alzheimer's disease. Effective therapeutic halting neurodegeneration, validated already in 5XFAD mice.
Collapse
Affiliation(s)
| | - María‐Salud García‐Ayllón
- Unidad de InvestigaciónHospital General Universitario de Elche, FISABIOElcheSpain
- Instituto de Neurociencias de AlicanteUniversidad Miguel Hernández‐CSICSant Joan d'AlacantSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain
| | - Neus Falgàs
- Alzheimer's disease and other cognitive disorders UnitHospital Clínic de Barcelona. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Sharon A. Brangman
- Department of GeriatricsUpstate Center of Excellence for Alzheimer's DiseaseSUNY Upstate Medical University 750 East Adams StreetSyracuseNew YorkUSA
| | - Margaret M Esiri
- Neuropathology DepartmentJohn Radcliffe Hospital, West WingOxford UniversityOxfordUK
| | - Clive W. Coen
- Faculty of Life Sciences & MedicineKing's College LondonLondonUK
| | | |
Collapse
|
28
|
Ali AM, Mohamed AA, Ibrahim AN, Elfiky AA. Acetylcholinesterase - glucose-regulated protein 78 binding site prediction, a hope to cure neurological disorders such as Alzheimer's disease. J Recept Signal Transduct Res 2024; 44:122-128. [PMID: 39522163 DOI: 10.1080/10799893.2024.2426523] [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: 05/25/2024] [Revised: 10/24/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Cerebral amyloid plaques in the brain define the elderly neuralgic disorder, Alzheimer's disease (AD). The enzyme Acetylcholinesterase (AChE) was reported to play a vital role in AD. It was shown that AChE induces amyloid fibril formation forming highly toxic AChE-Amyloid-β (Aβ) complexes. AChE can accelerate amyloid formation, and its inhibition could prevent such alterations to the enzyme. Understanding the proteostasis of AChE and its binding site to cellular chaperone GRP78 (Glucose-regulated protein 78) would help find a treatment for AD. In this study, the state of the art computational tools were utilized to predict the binding location of AChE that can stably associate with the cellular chaperone, GRP78. Sequence comparison along with molecular docking predicts two binding locations on AChE (C69-C96 and C257-C272) that could bind to GRP78 substrate binding domain β (SBDβ). The analysis of the docking data suggests that the former location has the best average binding affinity value (-12.16 kcal/mol) and average interaction pattern (13.9 ± 3.5 H-bonds, 5.5 ± 1.4 hydrophobic contacts, and 1.4 ± 1.2 salt bridges).
Collapse
Affiliation(s)
- Ahmed M Ali
- Biotechnology Department, Cairo University, Giza, Egypt
| | | | | | - Abdo A Elfiky
- Biophysics Department, Cairo University, Giza, Egypt
| |
Collapse
|
29
|
Pourfridoni M, Hedayati-Moghadam M, Fathi S, Fathi S, Mirrashidi FS, Askarpour H, Shafieemojaz H, Baghcheghi Y. Beneficial effects of metformin treatment on memory impairment. Mol Biol Rep 2024; 51:640. [PMID: 38727848 DOI: 10.1007/s11033-024-09445-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/13/2024] [Indexed: 07/12/2024]
Abstract
Memory issues are a prevalent symptom in different neurodegenerative diseases and can also manifest in certain psychiatric conditions. Despite limited medications approved for treating memory problems, research suggests a lack of sufficient options in the market. Studies indicate that a significant percentage of elderly individuals experience various forms of memory disorders. Metformin, commonly prescribed for type 2 diabetes, has shown neuroprotective properties through diverse mechanisms. This study explores the potential of metformin in addressing memory impairments. The current research gathered its data by conducting an extensive search across electronic databases including PubMed, Web of Science, Scopus, and Google Scholar. Previous research suggests that metformin enhances brain cell survival and memory function in both animal and clinical models by reducing oxidative stress, inflammation, and cell death while increasing beneficial neurotrophic factors. The findings of the research revealed that metformin is an effective medication for enhancing various types of memory problems in numerous studies. Given the rising incidence of memory disorders, it is plausible to utilize metformin, which is an affordable and accessible drug. It is often recommended as a treatment to boost memory.
Collapse
Affiliation(s)
- Mohammad Pourfridoni
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
- Clinical Research Development Center of Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Mahdiyeh Hedayati-Moghadam
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
- Clinical Research Development Center of Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Shirin Fathi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- Clinical Research Development Center of Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Shiva Fathi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Clinical Research Development Center of Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fatemeh Sadat Mirrashidi
- Departrment of Pediatrics, Jiroft University of Medical Sciences, Jiroft, Iran
- Clinical Research Development Center of Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Hedyeh Askarpour
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
- Clinical Research Development Center of Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Hadi Shafieemojaz
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
- Clinical Research Development Center of Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Yousef Baghcheghi
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran.
- Clinical Research Development Center of Imam Khomeini Hospital, Jiroft University of Medical Sciences, Jiroft, Iran.
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
30
|
Vasantharekha R, Priyanka HP, Nair RS, Hima L, Pratap UP, Srinivasan AV, ThyagaRajan S. Alterations in Immune Responses Are Associated with Dysfunctional Intracellular Signaling in Peripheral Blood Mononuclear Cells of Men and Women with Mild Cognitive Impairment and Alzheimer's disease. Mol Neurobiol 2024; 61:2964-2977. [PMID: 37957423 DOI: 10.1007/s12035-023-03764-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023]
Abstract
Deficits in the neuroendocrine-immune network in the periphery associated with the onset and progression of mild cognitive impairment (MCI) and Alzheimer's disease (AD) have not been extensively studied. The present study correlatively examines the association between cell-mediated immune responses, stress hormones, amyloid precursor protein (APP) expression, peripheral blood mononuclear cells (PBMC), and intracellular signaling molecules in the pathophysiology of MCI and AD compared to adults. Serum APP, lymphocyte proliferation, total cholinesterase (TChE), butyrylcholinesterase (BChE) activities, cytokines (IL-2, IFN-γ, IL-6, and TNF-α), and intracellular signaling molecules (p-ERK, p-CREB, and p-Akt) were measured in the PBMCs of adult, old, MCI, and AD men and women initially and after 3 years in the same population. An age- and disease-associated decline in mini-mental state examination (MMSE) scores and lymphocyte proliferation of MCI and AD men and women were observed. An age- and disease-related increase in serum APP, cortisol levels, and TChE activity were observed in men and women. Enhanced production of Th1 cytokine, IL-2, pro-inflammatory cytokines, and suppressed intracellular transcription factors may promote the inflammatory environment in MCI and AD patients. The expression of CREB and Akt was lower in MCI and AD men, while the expression of p-ERK was higher, and p-CREB was lower in MCI and AD women after 3 years. These results suggest that changes in specific intracellular signaling pathways may influence alterations in cell-mediated immunity to promote disease progression in MCI and AD patients.
Collapse
Affiliation(s)
- Ramasamy Vasantharekha
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science & Technology, Kattankulathur, 603203, Tamil Nadu, India.
| | - Hannah P Priyanka
- Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai, Tamil Nadu, India
| | - Rahul S Nair
- Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai, Tamil Nadu, India
| | - Lalgi Hima
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science & Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Uday P Pratap
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science & Technology, Kattankulathur, 603203, Tamil Nadu, India
| | | | - Srinivasan ThyagaRajan
- Integrative Medicine Laboratory, Department of Biotechnology, SRM Institute of Science & Technology, Kattankulathur, 603203, Tamil Nadu, India
| |
Collapse
|
31
|
Nazir S. Salivary biomarkers: The early diagnosis of Alzheimer's disease. Aging Med (Milton) 2024; 7:202-213. [PMID: 38725701 PMCID: PMC11077336 DOI: 10.1002/agm2.12282] [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: 10/07/2023] [Revised: 10/25/2023] [Accepted: 12/19/2023] [Indexed: 05/12/2024] Open
Abstract
The precise identification of Alzheimer's disease and other prevalent neurodegenerative diseases remains a difficult issue that requires the development of early detection of the disease and inexpensive biomarkers that can replace the present cerebrospinal fluid and imaging biomarkers. Blood biomarkers, such as amyloid and neurofilament light, have been emphasized as an important and practical tool in a testing or examination procedure thanks to advancements in ultra-sensitive detection techniques. Although saliva is not currently being researched for neurodegenerative diseases, it is an important source of biomarkers that can be used for the identification of diseases and has some advantages over other biofluids. While this may be true for most people, getting saliva from elderly people presents some significant challenges. In this overview, we will first discuss how saliva is created and how aging-related illnesses may affect the amount and kind of saliva produced. The findings support the use of salivary amyloid protein, tau species, and novel biomarkers in the diagnosis of Alzheimer's disease.
Collapse
Affiliation(s)
- Sophia Nazir
- Wolfson Nanomaterials and Devices Laboratory, School of Computing, Electronics and MathematicsPlymouth UniversityDevonUK
| |
Collapse
|
32
|
Sinha JK, Trisal A, Ghosh S, Gupta S, Singh KK, Han SS, Mahapatra M, Abomughaid MM, Abomughayedh AM, Almutary AG, Iqbal D, Bhaskar R, Mishra PC, Jha SK, Jha NK, Singh AK. Psychedelics for alzheimer's disease-related dementia: Unveiling therapeutic possibilities and pathways. Ageing Res Rev 2024; 96:102211. [PMID: 38307424 DOI: 10.1016/j.arr.2024.102211] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024]
Abstract
Psychedelics have traditionally been used for spiritual and recreational purposes, but recent developments in psychotherapy have highlighted their potential as therapeutic agents. These compounds, which act as potent 5-hydroxytryptamine (5HT) agonists, have been recognized for their ability to enhance neural plasticity through the activation of the serotoninergic and glutamatergic systems. However, the implications of these findings for the treatment of neurodegenerative disorders, particularly dementia, have not been fully explored. In recent years, studies have revealed the modulatory and beneficial effects of psychedelics in the context of dementia, specifically Alzheimer's disease (AD)-related dementia, which lacks a definitive cure. Psychedelics such as N,N-dimethyltryptamine (DMT), lysergic acid diethylamide (LSD), and Psilocybin have shown potential in mitigating the effects of this debilitating disease. These compounds not only target neurotransmitter imbalances but also act at the molecular level to modulate signalling pathways in AD, including the brain-derived neurotrophic factor signalling pathway and the subsequent activation of mammalian target of rapamycin and other autophagy regulators. Therefore, the controlled and dose-dependent administration of psychedelics represents a novel therapeutic intervention worth exploring and considering for the development of drugs for the treatment of AD-related dementia. In this article, we critically examined the literature that sheds light on the therapeutic possibilities and pathways of psychedelics for AD-related dementia. While this emerging field of research holds great promise, further studies are necessary to elucidate the long-term safety, efficacy, and optimal treatment protocols. Ultimately, the integration of psychedelics into the current treatment paradigm may provide a transformative approach for addressing the unmet needs of individuals living with AD-related dementia and their caregivers.
Collapse
Affiliation(s)
| | - Anchal Trisal
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra 411057, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, the Republic of Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, the Republic of Korea
| | | | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ali M Abomughayedh
- Pharmacy Department, Aseer Central Hospital, Ministry of Health, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, the Republic of Korea; Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, the Republic of Korea.
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, India.
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
33
|
Jiao LL, Dong HL, Liu MM, Wu PL, Cao Y, Zhang Y, Gao FG, Zhu HY. The potential roles of salivary biomarkers in neurodegenerative diseases. Neurobiol Dis 2024; 193:106442. [PMID: 38382884 DOI: 10.1016/j.nbd.2024.106442] [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: 11/21/2023] [Revised: 02/01/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024] Open
Abstract
Current research efforts on neurodegenerative diseases are focused on identifying novel and reliable biomarkers for early diagnosis and insight into disease progression. Salivary analysis is gaining increasing interest as a promising source of biomarkers and matrices for measuring neurodegenerative diseases. Saliva collection offers multiple advantages over the currently detected biofluids as it is easily accessible, non-invasive, and repeatable, allowing early diagnosis and timely treatment of the diseases. Here, we review the existing findings on salivary biomarkers and address the potential value in diagnosing neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Based on the available research, β-amyloid, tau protein, α-synuclein, DJ-1, Huntington protein in saliva profiles display reliability and validity as the biomarkers of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ling-Ling Jiao
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China; School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Hui-Lin Dong
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Meng-Meng Liu
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Peng-Lin Wu
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Yi Cao
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Yuan Zhang
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Fu-Gao Gao
- Xuzhou Cigarette Factory, China Tobacco Jiangsu Industrial Co Ltd, Xuzhou 221005, China.
| | - Huai-Yuan Zhu
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China; School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| |
Collapse
|
34
|
Alhawarri MB, Al-Thiabat MG, Dubey A, Tufail A, Fouad D, Alrimawi BH, Dayoob M. ADME profiling, molecular docking, DFT, and MEP analysis reveal cissamaline, cissamanine, and cissamdine from Cissampelos capensis L.f. as potential anti-Alzheimer's agents. RSC Adv 2024; 14:9878-9891. [PMID: 38528929 PMCID: PMC10961956 DOI: 10.1039/d4ra01070a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 03/17/2024] [Indexed: 03/27/2024] Open
Abstract
The current pharmacotherapies for Alzheimer's disease (AD) demonstrate limited efficacy and are associated with various side effects, highlighting the need for novel therapeutic agents. Natural products, particularly from medicinal plants, have emerged as a significant source of potential neuroprotective compounds. In this context, Cissampelos capensis L.f., renowned for its medicinal properties, has recently yielded three new proaporphine alkaloids; cissamaline, cissamanine, and cissamdine. Despite their promising bioactive profiles, the biological targets of these alkaloids in the context of AD have remained unexplored. This study undertakes a comprehensive in silico examination of the binding affinity and molecular interactions of these alkaloids with human protein targets implicated in AD. The drug likeness and ADME analyses indicate favorable pharmacokinetic profiles for these compounds, suggesting their potential efficacy in targeting the central nervous system. Molecular docking studies indicate that cissamaline, cissamanine, and cissamdine interact with key AD-associated proteins. These interactions are comparable to, or in some aspects slightly less potent than, those observed with established AD drugs, highlighting their potential as novel therapeutic agents for Alzheimer's disease. Crucially, Density Functional Theory (DFT) calculations offer deep insights into the electronic and energetic characteristics of these alkaloids. These calculations reveal distinct electronic properties, with differences in total energy, binding energy, HOMO-LUMO gaps, dipole moments, and electrophilicity indices. Such variations suggest unique reactivity profiles and molecular stability, pertinent to their pharmacological potential. Moreover, Molecular Electrostatic Potential (MEP) analyses provide visual representations of the electrostatic characteristics of these alkaloids. The analyses highlight areas prone to electrophilic and nucleophilic attacks, indicating their potential for specific biochemical interactions. This combination of DFT and MEP results elucidates the intricate electronic, energetic, and electrostatic properties of these compounds, underpinning their promise as AD therapeutic agents. The in silico findings of this study shed light on the promising potential of cissamaline, cissamanine, and cissamdine as agents for AD treatment. However, further in vitro and in vivo studies are necessary to validate these theoretical predictions and to understand the precise mechanisms through which these alkaloids may exert their therapeutic effects.
Collapse
Affiliation(s)
- Maram B Alhawarri
- Department of Pharmacy, Faculty of Pharmacy, Jadara University P.O.Box 733 Irbid 21110 Jordan
| | - Mohammad G Al-Thiabat
- School of Pharmaceutical Sciences, Universiti Sains Malaysia Gelugor 11800 Penang Malaysia
| | - Amit Dubey
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences Chennai-600077 Tamil Nadu India
- Computational Chemistry and Drug Discovery Division Quanta Calculus Greater Noida-201310 Uttar Pradesh India
| | - Aisha Tufail
- Computational Chemistry and Drug Discovery Division Quanta Calculus Greater Noida-201310 Uttar Pradesh India
| | - Dania Fouad
- Faculty of Dentistry, Ibn Sina University for Medical and Pharmaceutical Sciences Baghdad Iraq
| | | | | |
Collapse
|
35
|
Ribeiro J, Araújo-Silva H, Fernandes M, da Silva JA, Pinto FDCL, Pessoa ODL, Santos HS, de Menezes JESA, Gomes AC. Petrosamine isolated from marine sponge Petrosia sp. demonstrates protection against neurotoxicity in vitro and in vivo. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:16. [PMID: 38383833 PMCID: PMC10881933 DOI: 10.1007/s13659-024-00439-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
According to The World Alzheimer Report 2023 by Alzheimer's Disease International (ADI) estimates that 33 to 38.5 million people worldwide suffer from Alzheimer's Disease (AD). A crucial hallmark associated with this disease is associated with the deficiency of the brain neurotransmitter acetylcholine, due to an affected acetylcholinesterase (AChE) activity. Marine organisms synthesize several classes of compounds, some of which exhibit significant AChE inhibition, such as petrosamine, a coloured pyridoacridine alkaloid. The aim of this work was to characterize the activity of petrosamine isolated for the first time from a Brazilian marine sponge, using two neurotoxicity models with aluminium chloride, as exposure to aluminium is associated with the development of neurodegenerative diseases. The in vitro model was based in a neuroblastoma cell line and the in vivo model exploited the potential of zebrafish (Danio rerio) embryos in mimicking hallmarks of AD. To our knowledge, this is the first report on petrosamine's activity over these parameters, either in vitro or in vivo, in order to characterize its full potential for tackling neurotoxicity.
Collapse
Affiliation(s)
- Joana Ribeiro
- CBMA (Centre of Molecular and Environmental Biology) / Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Henrique Araújo-Silva
- CBMA (Centre of Molecular and Environmental Biology) / Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Mário Fernandes
- CBMA (Centre of Molecular and Environmental Biology) / Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Joilna Alves da Silva
- Program in Natural Sciences, Natural Products Chemistry Laboratory, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Francisco das Chagas L Pinto
- Department of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Otília Deusdenia L Pessoa
- Department of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Hélcio Silva Santos
- Program in Natural Sciences, Natural Products Chemistry Laboratory, State University of Ceará, Fortaleza, Ceará, Brazil
- Department of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | - Andreia C Gomes
- CBMA (Centre of Molecular and Environmental Biology) / Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.
| |
Collapse
|
36
|
Sanfeliu C, Bartra C, Suñol C, Rodríguez-Farré E. New insights in animal models of neurotoxicity-induced neurodegeneration. Front Neurosci 2024; 17:1248727. [PMID: 38260026 PMCID: PMC10800989 DOI: 10.3389/fnins.2023.1248727] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
The high prevalence of neurodegenerative diseases is an unintended consequence of the high longevity of the population, together with the lack of effective preventive and therapeutic options. There is great pressure on preclinical research, and both old and new models of neurodegenerative diseases are required to increase the pipeline of new drugs for clinical testing. We review here the main models of neurotoxicity-based animal models leading to central neurodegeneration. Our main focus was on studying how changes in neurotransmission and neuroinflammation, mainly in rodent models, contribute to harmful processes linked to neurodegeneration. The majority of the models currently in use mimic Parkinson's disease (PD) and Alzheimer's disease (AD), which are the most common neurodegenerative conditions in older adults. AD is the most common age-related dementia, whereas PD is the most common movement disorder with also cases of dementia. Several natural toxins and xenobiotic agents induce dopaminergic neurodegeneration and can reproduce neuropathological traits of PD. The literature analysis of MPTP, 6-OH-dopamine, and rotenone models suggested the latter as a useful model when specific doses of rotenone were administrated systemically to C57BL/6 mice. Cholinergic neurodegeneration is mainly modelled with the toxin scopolamine, which is a useful rodent model for the screening of protective drugs against cognitive decline and AD. Several agents have been used to model neuroinflammation-based neurodegeneration and dementia in AD, including lipopolysaccharide (LPS), streptozotocin, and monomeric C-reactive protein. The bacterial agent LPS makes a useful rodent model for testing anti-inflammatory therapies to halt the development and severity of AD. However, neurotoxin models might be more useful than genetic models for drug discovery in PD but that is not the case in AD where they cannot beat the new developments in transgenic mouse models. Overall, we should work using all available models, either in vivo, in vitro, or in silico, considering the seriousness of the moment and urgency of developing effective drugs.
Collapse
Affiliation(s)
- Coral Sanfeliu
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Clara Bartra
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- PhD Program in Biotechnology, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Barcelona, Spain
| | - Cristina Suñol
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Eduard Rodríguez-Farré
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
37
|
Egbujor MC. Sulfonamide Derivatives: Recent Compounds with Potent Anti-alzheimer's Disease Activity. Cent Nerv Syst Agents Med Chem 2024; 24:82-104. [PMID: 38275073 DOI: 10.2174/0118715249278489231128042135] [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: 09/14/2023] [Revised: 10/27/2023] [Accepted: 11/06/2023] [Indexed: 01/27/2024]
Abstract
Facile synthetic procedures and broad spectrum of biological activities are special attributes of sulfonamides. Sulfonamide derivatives have demonstrated potential as a class of compounds for the treatment of Alzheimer's disease (AD). Recent sulfonamide derivatives have been reported as prospective anti-AD agents, with a focus on analogues that significantly inhibit the function of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) enzymes and exhibit remarkable antioxidant and anti-inflammatory properties, all of which are critical for the treatment of AD. Sulfonamide- mediated activation of nuclear factor erythroid 2-related factor 2 (NRF2), a key regulator of the endogenous antioxidant response, has also been suggested as a potential therapeutic approach in AD. Additionally, it has been discovered that a number of sulfonamide derivatives show selectivity for the β- and γ-secretase enzymes and a significant reduction of amyloid B (Aβ) aggregation, which have been implicated in AD. The comparative molecular docking of benzenesulfonamide and donepezil, an AD reference drug showed comparable anti-AD activities. These suggest that sulfonamide derivatives may represent a new class of drugs for the treatment of AD. Thus, the current review will focus on recent studies on the chemical synthesis and evaluation of the anti-AD properties, molecular docking, pharmacological profile, and structure-activity relationship (SAR) of sulfonamide derivatives, as well as their potential anti-AD mechanisms of action. This paper offers a thorough assessment of the state of the art in this field of study and emphasizes the potential of sulfonamide derivatives synthesized during the 2012-2023 period as a new class of compounds for the treatment of AD.
Collapse
|
38
|
de la Monte SM, Tong M. Agent Orange Herbicidal Toxin-Initiation of Alzheimer-Type Neurodegeneration. J Alzheimers Dis 2024; 97:1703-1726. [PMID: 38306038 PMCID: PMC10979462 DOI: 10.3233/jad-230881] [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] [Indexed: 02/03/2024]
Abstract
Background Agent Orange (AO) is a Vietnam War-era herbicide that contains a 1 : 1 ratio of 2,4-dichlorophenoxyacetic acid (2,4-D) and 2,4,5-trichlorophenoxyacetic acid (2,4,5-T). Emerging evidence suggests that AO exposures cause toxic and degenerative pathologies that may increase the risk for Alzheimer's disease (AD). Objective This study investigates the effects of the two main AO constituents on key molecular and biochemical indices of AD-type neurodegeneration. Methods Long Evans rat frontal lobe slice cultures treated with 250μg/ml of 2,4-D, 2,4,5-T, or both (D + T) were evaluated for cytotoxicity, oxidative injury, mitochondrial function, and AD biomarker expression. Results Treatment with the AO constituents caused histopathological changes corresponding to neuronal, white matter, and endothelial cell degeneration, and molecular/biochemical abnormalities indicative of cytotoxic injury, lipid peroxidation, DNA damage, and increased immunoreactivity to activated Caspase 3, glial fibrillary acidic protein, ubiquitin, tau, paired-helical filament phosphorylated tau, AβPP, Aβ, and choline acetyltransferase. Nearly all indices of cellular injury and degeneration were more pronounced in the D + T compared with 2,4-D or 2,4,5-T treated cultures. Conclusions Exposures to AO herbicidal chemicals damage frontal lobe brain tissue with molecular and biochemical abnormalities that mimic pathologies associated with early-stage AD-type neurodegeneration. Additional research is needed to evaluate the long-term effects of AO exposures in relation to aging and progressive neurodegeneration in Vietnam War Veterans.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology and Laboratory Medicine, Neurology, and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
39
|
Alamri OA, Qusti S, Balgoon M, Ageeli AA, Al-Marhaby FA, Alosaimi AM, Jowhari MA, Saeed A. The role of MoS 2 QDs coated with DSPE-PEG-TPP in the protection of protein secondary structure of the brain tissues in an Alzheimer's disease model. Int J Biol Macromol 2024; 255:128522. [PMID: 38040141 DOI: 10.1016/j.ijbiomac.2023.128522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/12/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
In this investigation, we have explored the protective capacity of MoS2 QDs coated with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethyleneglycol) -2000] (DSPE-PEG) linked with (3-carboxypropyl) triphenylphosphonium-bromide (TPP), on the secondary structure of proteins in Alzheimer's disease (AD)-affected brain tissues. Using a cohort of fifteen male SWR/J mice, we establish three groups: a control group, a second group induced with AD through daily doses of AlCl3 and D-galactose for 49 consecutive days, and a third group receiving the same AD-inducing doses but treated with DSPE-PEG-TPP-MoS2 QDs. Brain tissues are meticulously separated from the skull, and their molecular structures are analyzed via FTIR spectroscopy. Employing the curve fitting method on the amide I peak, we delve into the nuances of protein secondary structure. The FTIR analysis reveals a marked increase in β-sheet structures and a concurrent decline in turn and α-helix structures in the AD group in comparison to the control group. Notably, no statistically significant differences emerge between the treated and control mice. Furthermore, multivariate analysis of the FTIR spectral region, encompassing protein amide molecular structures, underscores a remarkable similarity between the treated and normal mice. This study elucidates the potential of DSPE-PEG-TPP-MoS2 QDs in shielding brain tissue proteins against the pathogenic influences of AD.
Collapse
Affiliation(s)
- Ohoud Abdulaziz Alamri
- Department of Medical Laboratory, King Fahad Armed Forces Hospital, Jeddah 23311, Saudi Arabia; Department of Biochemistry Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Safaa Qusti
- Department of Biochemistry Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maha Balgoon
- Department of Biochemistry Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abeer A Ageeli
- Department of Chemistry, Faculty of Science, Jazan University, Jazan 45142, Saudi Arabia
| | - F A Al-Marhaby
- Department of Physics, Al-Qunfudhah University College, Umm Al-Qura University, Makkah 24230, Saudi Arabia
| | - Abeer M Alosaimi
- Department of Chemistry, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Mohammed A Jowhari
- Medical Physics Department, Jazan Specialized Hospital, Ministry of Health, Jazan Health Affairs, Jazan 45142, Saudi Arabia
| | - Abdu Saeed
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Physics, Thamar University, Thamar 87246, Yemen.
| |
Collapse
|
40
|
Dassamiour S, Bensaad MS, Ghebache W. Utility of phenolic acids in neurological disorders. ADVANCEMENT OF PHENOLIC ACIDS IN DRUG DISCOVERY 2024:295-344. [DOI: 10.1016/b978-0-443-18538-0.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
41
|
Budhbhatti U, Chauhan A, Bhatt D, Parmar C, Damani V, Patel A, Joshi C. Association of NOTCH4 and ACHE gene polymorphism in Alzheimer's disease of Gujarat cohort. Neurosci Lett 2023; 814:137428. [PMID: 37544578 DOI: 10.1016/j.neulet.2023.137428] [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: 04/19/2023] [Revised: 07/03/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Alzheimer's Disease (AD) is the most common form of dementia, affecting cognitive and behavioral functions. AD is a complex disease resulting from the modest effect of gene interaction and environmental factors, as a result of which the exact pathogenesis is still unknown. AIM The aim of the present study was to investigate the association between variants of 98 targeted genes with Alzheimer's disease phenotype. METHOD A total of 98 genes from 32 AD cases and 11 controls were genotyped using the Haloplex target enrichment method and the PCR-RFLP approach.Association analysis was performed using the PLINK tool to identify the variant significantly associated with AD. Functional enrichment analysis and network analysis was performed using ClueGo and String database respectively. The Expression Quantitative Trait Loci (eQTL) analysis using the Genotype Tissue Expression (GTEx) dataset to explore the possible implication of the variant on the expression of one or more genes in different brain regions and whole blood. RESULT Association analysis showed significant association of 19 variant assigned to 16 genes with Alzheimer's with p-value < 0.05 with rs367398/NOTCH4 only variant that passed multiple test corrections. Functional enrichment analysis showed association of these genes with AD. ClueGo and network analysis utilizing the String database suggested that genes are directly and indirectly linked to the AD pathogenesis. eQTL analysis revealed that the rs367398/NOTCH4 and rs1799806/ACHE variant showed significant eQTL for the neighbouring genes. CONCLUSION The present study showed the possible role of 16 genes in AD pathogenesis, especially highlighting the role of rs367398/NOTCH4 and rs1799806/ACHE. However further investigation with large cohort is required to study and validate the implication of these variants in the AD pathogenesis.
Collapse
Affiliation(s)
- Urvi Budhbhatti
- Gujarat Biotechnology Research Centre (GBRC), Gandhinagar, Department of Science and Technology, Government of Gujarat, India
| | - Ajay Chauhan
- Hospital of Mental Health-Gujarat Institute of Mental Health, Shahibaug, Ahmedabad, Gujarat, India
| | - Deeptiben Bhatt
- Hospital of Mental Health-Gujarat Institute of Mental Health, Shahibaug, Ahmedabad, Gujarat, India
| | - Chirag Parmar
- Hospital of Mental Health-Gujarat Institute of Mental Health, Shahibaug, Ahmedabad, Gujarat, India
| | - Vishalbhai Damani
- Hospital of Mental Health-Gujarat Institute of Mental Health, Shahibaug, Ahmedabad, Gujarat, India
| | - Amrutlal Patel
- Gujarat Biotechnology Research Centre (GBRC), Gandhinagar, Department of Science and Technology, Government of Gujarat, India.
| | - Chaitanya Joshi
- Gujarat Biotechnology Research Centre (GBRC), Gandhinagar, Department of Science and Technology, Government of Gujarat, India.
| |
Collapse
|
42
|
Wang L, Sun T, Wang Z, Liu H, Qiu W, Tang X, Guo H, Yang P, Chen Y, Sun H. Design, Synthesis, and Proof of Concept of Balanced Dual Inhibitors of Butyrylcholinesterase (BChE) and Histone Deacetylase 6 (HDAC6) for the Treatment of Alzheimer's Disease. ACS Chem Neurosci 2023; 14:3226-3248. [PMID: 37561893 DOI: 10.1021/acschemneuro.3c00358] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023] Open
Abstract
Concomitant inhibition of butyrylcholinesterase (BChE) and histone deacetylase 6 (HDAC6) is supposed to be effective in the treatment of Alzheimer's disease (AD). Inspired by our previous efforts in designing BChE inhibitors, herein, selective BChE and HDAC6 dual inhibitors were successfully identified through the fusion of the core pharmacophoric moiety of BChE and HDAC6 inhibitors. After the structure-activity relationship (SAR) studies, two compounds (24g and 29a) were confirmed to have superior inhibitory activity against BChE (the IC50 against hBChE are 4.0 and 1.8 nM, respectively) and HDAC6 (the IC50 against HDAC6 are 8.9 and 71.0 nM, respectively). These two compounds showed prominently neuroprotective effects in vitro, potent reactive oxygen species (ROS) scavenging effects, and effective metal ion (Fe2+ and Cu2+) chelation. In addition, they exhibited pronounced inhibition of phosphorylated tau and a moderate immunomodulatory effect, with a lack of neurotoxicity at the cellular level. In vivo studies showed that both 24g and 29a ameliorated the cognitive impairment in an Aβ1-42-induced mouse model at a low dosage (2.5 mg/kg). Our data demonstrated that BChE/HDAC6 dual inhibitors could establish the basis for a potential new symptomatic and disease-modifying strategy to treat AD.
Collapse
Affiliation(s)
- Lei Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Tianyu Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Zhenqi Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Hui Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Weimin Qiu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Xu Tang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Huanchao Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Peng Yang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
43
|
Basli A, Bounaas J. Pathophysiological mechanism and natural preventive and therapeutic strategies of Alzheimer's disease. Nutr Health 2023; 29:403-413. [PMID: 36377316 DOI: 10.1177/02601060221137104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) is characterized by the presence of two types of protein deposits in the brain, amyloid plaques and neurofibrillary tangles. The first one are dense deposits of beta amyloid protein, the second one are dense deposits of the protein tau. These proteins are present in all of our brains, but in AD they act unusually, leading to neuronal degeneration. This review will provide an overview of the AD, including the role of amyloid beta and tau, and mechanisms that lead to the formation of plaques and tangles. The review will also cover the existing researches that have focused on the inhibition of amyloid beta formation, cholinesterase, tau hyperphosphorylation, the pathogenic mechanisms of apoE4, and GSK-3 as a solution that could be used to slow or prevent the disease.
Collapse
Affiliation(s)
- Abdelkader Basli
- Laboratory of Interaction Research, Biodiversity, Ecosystems and Biotechnology, Faculty of Sciences, University of Skikda, Skikda, Algeria
| | - Jihane Bounaas
- Laboratory of Interaction Research, Biodiversity, Ecosystems and Biotechnology, Faculty of Sciences, University of Skikda, Skikda, Algeria
| |
Collapse
|
44
|
de la Monte SM, Goel A, Tong M, Delikkaya B. Agent Orange Causes Metabolic Dysfunction and Molecular Pathology Reminiscent of Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:751-766. [PMID: 37662613 PMCID: PMC10473158 DOI: 10.3233/adr-230046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 09/05/2023] Open
Abstract
Background Agent Orange, an herbicide used during the Vietnam War, contains 2,4-dichlorophenoxyacetic acid (2,4-D) and 2,4,5-trichlorophenoxyacetic acid (2,4,5-T). Agent Orange has teratogenic and carcinogenic effects, and population-based studies suggest Agent Orange exposures lead to higher rates of toxic and degenerative pathologies in the peripheral and central nervous system (CNS). Objective This study examines the potential contribution of Agent Orange exposures to neurodegeneration. Methods Human CNS-derived neuroepithelial cells (PNET2) treated with 2,4-D and 2,4,5-T were evaluated for viability, mitochondrial function, and Alzheimer's disease (AD)-related proteins. Results Treatment with 250μg/ml 2,4-D or 2,4,5-T significantly impaired mitochondrial function, caused degenerative morphological changes, and reduced viability in PNET2 cells. Correspondingly, glyceraldehyde-3-phosphate dehydrogenase expression which is insulin-regulated and marks the integrity of carbohydrate metabolism, was significantly inhibited while 4-hydroxy-2-nonenal, a marker of lipid peroxidation, was increased. Tau neuronal cytoskeletal protein was significantly reduced by 2,4,5-T, and relative tau phosphorylation was progressively elevated by 2,4,5-T followed by 2,4-D treatment relative to control. Amyloid-β protein precursor (AβPP) was increased by 2,4,5-T and 2,4-D, and 2,4,5-T caused a statistical trend (0.05 < p<0.10) increase in Aβ. Finally, altered cholinergic function due to 2,4,5-T and 2,4-D exposures was marked by significantly increased choline acetyltransferase and decreased acetylcholinesterase expression, corresponding with responses in early-stage AD. Conclusion Exposures to Agent Orange herbicidal chemicals rapidly damage CNS neurons, initiating a path toward AD-type neurodegeneration. Additional research is needed to understand the permanency of these neuropathologic processes and the added risks of developing AD in Agent Orange-exposed aging Vietnam Veterans.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and The Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Neurology and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Anuva Goel
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Busra Delikkaya
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
45
|
Uliassi E, Bergamini C, Rizzardi N, Naldi M, Cores Á, Bartolini M, Carlos Menéndez J, Bolognesi ML. Quinolinetrione-tacrine hybrids as multi-target-directed ligands against Alzheimer's disease. Bioorg Med Chem 2023; 91:117419. [PMID: 37487339 DOI: 10.1016/j.bmc.2023.117419] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/04/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023]
Abstract
Multi-target drug discovery is one of the most active fields in the search for new drugs against Alzheimer's disease (AD). This is because the complexity of AD pathological network might be adequately tackled by multi-target-directed ligands (MTDLs) aimed at modulating simultaneously multiple targets of such a network. In a continuation of our efforts to develop MTDLs for AD, we have been focusing on the molecular hybridization of the acetylcholinesterase inhibitor tacrine with the aim of expanding its anti-AD profile. Herein, we manipulated the structure of a previously developed tacrine-quinone hybrid (1). We designed and synthesized a novel set of MTDLs (2-6) by replacing the naphthoquinone scaffold of 1 with that of 2,5,8-quinolinetrione. The most interesting hybrid 3 inhibited cholinesterase enzymes at nanomolar concentrations. In addition, 3 exerted antioxidant effects in menadione-induced oxidative stress of SH-SY5Y cells. Importantly, 3 also showed low hepatotoxicity and good anti-amyloid aggregation properties. Remarkably, we uncovered the potential of the quinolinetrione scaffold, as a novel anti-amyloid aggregation and antioxidant motif to be used in further anti-AD MTDL drug discovery endeavors.
Collapse
Affiliation(s)
- Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Nicola Rizzardi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Marina Naldi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Ángel Cores
- Department of Chemistry in Pharmaceutical Sciences, Organic and Medicinal Chemistry Unit, Faculty of Pharmacy, Universidad Complutense, 28040 Madrid, Spain
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - J Carlos Menéndez
- Department of Chemistry in Pharmaceutical Sciences, Organic and Medicinal Chemistry Unit, Faculty of Pharmacy, Universidad Complutense, 28040 Madrid, Spain.
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
46
|
Ahmed Khan D, Shahid A, Sherif AE, Aati HY, Abdullah M, Mehmood K, Hussain M, Basit A, Ahmad Ghalloo B, ur Rehman Khan K. A detailed biochemical characterization, toxicological assessment and molecular docking studies of Launaea fragilis: An important medicinal xero-halophyte. Saudi Pharm J 2023; 31:1047-1060. [PMID: 37250362 PMCID: PMC10212792 DOI: 10.1016/j.jsps.2023.04.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
Launaea fragilis (Asso) Pau (Family: Asteraceae) is a wild medicinal plant that has been used in the folklore as a potential treatment for numerous ailments such as skin diseases, diarrhea, infected wounds, inflammation, child fever and hepatic pain. This study explored the chemical constitution, in-vivo toxicity, antimicrobial, antioxidant, and enzyme inhibition potential of ethanolic extract of L. fragilis (EELF). Additionally, in-silico docking studies of predominant compounds were performed against in-vitro tested enzymes. Similarly, in-silico ADMET properties of the compounds were performed to determine their pharmacokinetics, physicochemical properties, and toxicity profiles. The EELF was found rich in TFC (73.45 ± 0.25 mg QE/g) and TPC (109.02 ± 0.23 mg GAE/g). GC-MS profiling of EELF indicated the presence of a total of 47 compounds mainly fatty acids and essential oil. EELF showed no toxicity or growth retardation in chicks up to 300 mg/kg with no effect on the biochemistry and hematology of the chicks. EELF gave promising antioxidant activity through the CUPRAC method with an IC50 value of 13.14 ± 0.18 µg/ml. The highest inhibition activity against tyrosinase followed by acetylcholinesterase and α-Glucosidase was detected. Similarly, the antimicrobial study revealed the extract with good antibacterial and antiviral activity. A good docking score was observed in the in silico computational study of the predominant compounds. The findings revealed L. fragilis as a biocompatible, potent therapeutic alternative and suggest isolation and further in vivo pharmacological studies.
Collapse
Affiliation(s)
- Duraiz Ahmed Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur 63100, Punjab, Pakistan
| | - Afia Shahid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur 63100, Punjab, Pakistan
| | - Asmaa E. Sherif
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Alkharj 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Hanan Y. Aati
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Muhammad Abdullah
- Cholistan Institute of Desert Studies, The Islamia University of Bahawalpur 63100, Punjab, Pakistan
| | - Khalid Mehmood
- Department of Clinical Medicine and Surgery, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur 63100, Punjab, Pakistan
| | - Musaddique Hussain
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur 63100, Punjab, Pakistan
| | - Abdul Basit
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand
- Drug Delivery System Excellence Center, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand
| | - Bilal Ahmad Ghalloo
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur 63100, Punjab, Pakistan
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota 55454, MN, USA
| | - Kashif ur Rehman Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur 63100, Punjab, Pakistan
| |
Collapse
|
47
|
Nallasamy P, Rajamohamed BS, Jeyaraman J, Kathirvel B, Natarajan S. Regenerative marine waste towards CaCO 3 nanoformulation for Alzheimer's therapy. ENVIRONMENTAL RESEARCH 2023; 225:115631. [PMID: 36889568 DOI: 10.1016/j.envres.2023.115631] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/04/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
Alzheimer's disorder (AD) is associated with behavioural and cognitive destruction with due respect to the neurological degeneration. Conventional therapeutic approach for treatment of AD using neuroprotective drugs suffered certain limitations such as poor solubility, insufficient bioavailability, adverse side effects at higher dose and ineffective permeability on blood brain barrier (BBB). Development of nanomaterial based drug delivery system helped to overcome these barriers. Hence the present work focused on encapsulating neuroprotective drug citronellyl acetate within CaCO3 nanoparticles to develop neuroprotective CaCO3 nanoformulation (CA@CaCO3 NFs). CaCO3 was derived from marine conch shell waste, while the neuroprotective drug citronellyl acetate was scrutinized by in-silico high throughput screening. In-vitro findings revealed that CA@CaCO3 nanoformulation exhibited enhanced free radical scavenging activity of 92% (IC50 value - 29.27 ± 2.6 μg/ml), AChE inhibition of 95% (IC50 value - 25.6292 ± 1.5 μg/ml) at its maximum dose (100 μg/ml). CA@CaCO3 NFs attenuated the aggregation of β-amyloid peptide (Aβ) and also disaggregated the preformed mature plaques the major risk factor for AD. Overall, the present study reveals that CaCO3 nanoformulations exhibits potent neuroprotective potential when compared to the CaCO3 nanoparticles alone and citronellyl acetate alone due to the sustained drug release and synergistic effect of CaCO3 nanoparticles and citronellyl acetate depicting the fact that CaCO3 can act as promising drug delivery system for treatment of neurodegenerative and CNS related disorders.
Collapse
Affiliation(s)
- Prakashkumar Nallasamy
- Bionanomaterials Research Lab, Department of Nanoscience and Technology, Alagappa University, Tamilnadu, India
| | | | | | - Brindhadevi Kathirvel
- Center for Transdisciplinary Research (CFTR), Department of Pharmacology, Saveethe Dental College, Saveetha Institute of Medical and Technical Sciences, Saveeth University, Chennai, India
| | - Suganthy Natarajan
- Bionanomaterials Research Lab, Department of Nanoscience and Technology, Alagappa University, Tamilnadu, India.
| |
Collapse
|
48
|
Asproni B, Catto M, Loriga G, Murineddu G, Corona P, Purgatorio R, Cichero E, Fossa P, Scarano N, Martínez AL, Brea J, Pinna GA. Novel thienocycloalkylpyridazinones as useful scaffolds for acetylcholinesterase inhibition and serotonin 5-HT6 receptor interaction. Bioorg Med Chem 2023; 84:117256. [PMID: 37003157 DOI: 10.1016/j.bmc.2023.117256] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023]
Abstract
A library of eighteen thienocycloalkylpyridazinones was synthesized for human acetylcholinesterase (hAChE) and butyrylcholinesterase (hBChE) inhibition and serotonin 5-HT6 receptor subtype interaction by following a multitarget-directed ligand approach (MTDL), as a suitable strategy for treatment of Alzheimer's disease (AD). The novel compounds featured a tricyclic scaffold, namely thieno[3,2-h]cinnolinone, thienocyclopentapyridazinone and thienocycloheptapyridazinone, connected through alkyl chains of variable length to proper amine moieties, most often represented by N-benzylpiperazine or 1-(phenylsulfonyl)-4-(piperazin-1-ylmethyl)-1H-indole as structural elements addressing AChE and 5-HT6 interaction, respectively. Our study highlighted the versatility of thienocycloalkylpyridazinones as useful architectures for AChE interaction, with several N-benzylpiperazine-based analogues emerging as potent and selective hAChE inhibitors with IC50 in the 0.17-1.23 μM range, exhibiting low to poor activity for hBChE (IC50 = 4.13-9.70 μM). The introduction of 5-HT6 structural moiety phenylsulfonylindole in place of N-benzylpiperazine, in tandem with a pentamethylene linker, gave potent 5-HT6 thieno[3,2-h]cinnolinone and thienocyclopentapyridazinone-based ligands both displaying hAChE inhibition in the low micromolar range and unappreciable activity towards hBChE. While docking studies provided a rational structural explanation for AChE/BChE enzyme and 5-HT6 receptor interaction, in silico prediction of ADME properties of tested compounds suggested further optimization for development of such compounds in the field of MTDL for AD.
Collapse
|
49
|
Espinosa-Jiménez T, Cano A, Sánchez-López E, Olloquequi J, Folch J, Bulló M, Verdaguer E, Auladell C, Pont C, Muñoz-Torrero D, Parcerisas A, Camins A, Ettcheto M. A novel rhein-huprine hybrid ameliorates disease-modifying properties in preclinical mice model of Alzheimer's disease exacerbated with high fat diet. Cell Biosci 2023; 13:52. [PMID: 36895036 PMCID: PMC9999531 DOI: 10.1186/s13578-023-01000-y] [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: 12/29/2022] [Accepted: 02/28/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by a polyetiological origin. Despite the global burden of AD and the advances made in AD drug research and development, the cure of the disease remains elusive, since any developed drug has demonstrated effectiveness to cure AD. Strikingly, an increasing number of studies indicate a linkage between AD and type 2 diabetes mellitus (T2DM), as both diseases share some common pathophysiological features. In fact, β-secretase (BACE1) and acetylcholinesterase (AChE), two enzymes involved in both conditions, have been considered promising targets for both pathologies. In this regard, due to the multifactorial origin of these diseases, current research efforts are focusing on the development of multi-target drugs as a very promising option to derive effective treatments for both conditions. In the present study, we evaluated the effect of rhein-huprine hybrid (RHE-HUP), a synthesized BACE1 and AChE inhibitor, both considered key factors not only in AD but also in metabolic pathologies. Thus, the aim of this study is to evaluate the effects of this compound in APP/PS1 female mice, a well-established familial AD mouse model, challenged by high-fat diet (HFD) consumption to concomitantly simulate a T2DM-like condition. RESULTS Intraperitoneal treatment with RHE-HUP in APP/PS1 mice for 4 weeks reduced the main hallmarks of AD, including Tau hyperphosphorylation, Aβ42 peptide levels and plaque formation. Moreover, we found a decreased inflammatory response together with an increase in different synaptic proteins, such as drebrin 1 (DBN1) or synaptophysin, and in neurotrophic factors, especially in BDNF levels, correlated with a recovery in the number of dendritic spines, which resulted in memory improvement. Notably, the improvement observed in this model can be attributed directly to a protein regulation at central level, since no peripheral modification of those alterations induced by HFD consumption was observed. CONCLUSIONS Our results suggest that RHE-HUP could be a new candidate for the treatment of AD, even for individuals with high risk due to peripheral metabolic disturbances, given its multi-target profile which allows for the improvement of some of the most important hallmarks of the disease.
Collapse
Affiliation(s)
- Triana Espinosa-Jiménez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain.,Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Amanda Cano
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Ace Alzheimer Center Barcelona-International University of Catalunya (UIC), Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, Barcelona, Spain
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, Barcelona, Spain.,Unit of Synthesis and Biomedical Applications of Peptides, IQAC-CSIC, 08034, Barcelona, Spain
| | - Jordi Olloquequi
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Talca, Chile
| | - Jaume Folch
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Institut d'Investigació Sanitària Pere Virgili (IISPV), 43201, Reus, Spain.,Nutrition and Metabolic Health Research Group, Institute of Health Pere Virgili-IISPV, 43201, Reus, Spain
| | - Mònica Bulló
- Institut d'Investigació Sanitària Pere Virgili (IISPV), 43201, Reus, Spain.,Nutrition and Metabolic Health Research Group, Institute of Health Pere Virgili-IISPV, 43201, Reus, Spain.,CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029, Madrid, Spain
| | - Ester Verdaguer
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Carme Auladell
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Caterina Pont
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Diego Muñoz-Torrero
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.,Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Antoni Parcerisas
- Department of Basic Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain.,Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain. .,Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain. .,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain. .,Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Av. Joan XXIII 27/31, 08028, Barcelona, Spain.
| |
Collapse
|
50
|
Elsbaey M, Igarashi Y, Ibrahim MAA, Elattar E. Click-designed vanilloid-triazole conjugates as dual inhibitors of AChE and Aβ aggregation. RSC Adv 2023; 13:2871-2883. [PMID: 36756452 PMCID: PMC9850456 DOI: 10.1039/d2ra07539c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Based on their reported neuroprotective properties, vanilloids provide a good starting point for the synthesis of anti-Alzheimer's disease (AD) agents. In this context, nine new 1,2,3-triazole conjugates of vanilloids were synthesized via click chemistry. The compounds were tested for their effect on acetylcholine esterase (AChE) and amyloid-beta peptide (Aβ) aggregation. The triazole esters (E)-(1-(4-hydroxy-3-methoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl 3-(4-hydroxy-3 methoxyphenyl)acrylate 9 and (1-(4-hydroxy-3-methoxybenzyl)-1H-1,2,3-triazol-4-yl)methyl-4-hydroxy-3-methoxybenzoate 8 displayed dual inhibitory activity for AChE and Aβ aggregation with IC50 values of 0.47/0.31 μM and 1.2/0.95 μM, respectively, as compared to donepezil (0.27 μM) and tacrine (0.41 μM), respectively. The results showed that the triazole ester moiety is more favorable for the activity than the triazole ether moiety. This could be attributed to the longer length of the spacer between the two vanillyl moieties in the triazole esters. Furthermore, the binding affinities and modes of the triazole esters 9 and 8 were examined against AChE and Aβ utilizing a combination of docking predictions and molecular dynamics (MD) simulations. Docking computations revealed promising binding affinity of triazole esters 9 and 8 as potential AChE, Aβ40, and Aβ42 inhibitors with docking scores of -10.4 and -9.4 kcal mol-1, -5.8 and -4.7 kcal mol-1, and -3.3 and -2.9 kcal mol-1, respectively. The stability and binding energies of triazole esters 9 and 8 complexed with AChE, Aβ40, and Aβ42 were measured and compared to donepezil and tacrine over 100 ns MD simulations. According to the estimated binding energies, compounds 9 and 8 displayed good binding affinities with AChE, Aβ42, and Aβ40 with average ΔG binding values of -32.9 and -31.8 kcal mol-1, -12.0 and -10.5 kcal mol-1, and -20.4 and -16.6 kcal mol-1, respectively. Post-MD analyses demonstrated high steadiness for compounds 9 and 8 with AChE and Aβ during the 100 ns MD course. This work suggests the triazole conjugate of vanilloids as a promising skeleton for developing multi-target potential AD therapeutics.
Collapse
Affiliation(s)
- Marwa Elsbaey
- Pharmacognosy Department, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| | - Yasuhiro Igarashi
- Biotechnology Research Center and Department of Biotechnology, Toyama Prefectural University 5180 Kurokawa, Imizu Toyama 939-0398 Japan
| | - Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University 61519 Egypt
- School of Health Sciences, University of KwaZulu-Natal Westville Durban 4000 South Africa
| | - Eman Elattar
- Pharmacognosy Department, Faculty of Pharmacy, Mansoura University Mansoura 35516 Egypt
| |
Collapse
|