1
|
Mukherjee AG, Mishra S, Gopalakrishnan AV, Kannampuzha S, Murali R, Wanjari UR, B S, Vellingiri B, Madhyastha H, Kanagavel D, Vijayan M. Unraveling the mystery of citrate transporters in Alzheimer's disease: An updated review. Ageing Res Rev 2025; 107:102726. [PMID: 40073978 DOI: 10.1016/j.arr.2025.102726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/26/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
A key molecule in cellular metabolism, citrate is essential for lipid biosynthesis, energy production, and epigenetic control. The etiology of Alzheimer's disease (AD), a progressive neurodegenerative illness marked by memory loss and cognitive decline, may be linked to dysregulated citrate transport, according to recent research. Citrate transporters, which help citrate flow both inside and outside of cells, are becoming more and more recognized as possible participants in the molecular processes underlying AD. Citrate synthase (CS), a key enzyme in the tricarboxylic acid (TCA) cycle, supports mitochondrial function and neurotransmitter synthesis, particularly acetylcholine (ACh), essential for cognition. Changes in CS activity affect citrate availability, influencing energy metabolism and neurotransmitter production. Choline, a precursor for ACh, is crucial for neuronal function. Lipid metabolism, oxidative stress reactions, and mitochondrial function can all be affected by aberrant citrate transport, and these changes are linked to dementia. Furthermore, the two main pathogenic characteristics of AD, tau hyperphosphorylation and amyloid-beta (Aβ) aggregation, may be impacted by disturbances in citrate homeostasis. The goal of this review is to clarify the complex function of citrate transporters in AD and provide insight into how they contribute to the development and course of the illness. We aim to provide an in-depth idea of which particular transporters are dysregulated in AD and clarify the functional implications of these dysregulated transporters in brain cells. To reduce neurodegenerative processes and restore metabolic equilibrium, we have also discussed the therapeutic potential of regulating citrate transport. Gaining insight into the relationship between citrate transporters and the pathogenesis of AD may help identify new indicators for early detection and creative targets for treatment. This study offers hope for more potent ways to fight this debilitating illness and is a crucial step in understanding the metabolic foundations of AD.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Shatakshi Mishra
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Stany B
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab 151401, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan
| | - Deepankumar Kanagavel
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
2
|
Svobodova B, Moravcova Z, Misiachna A, Novakova G, Marek A, Finger V, Odvarkova J, Pejchal J, Karasova JZ, Netolicky J, Ladislav M, Hrabinova M, Sorf A, Muckova L, Fikejzlova L, Benkova M, Novak M, Prchal L, Capek J, Handl J, Rousar T, Greber KE, Ciura K, Horak M, Soukup O, Korabecny J. Novel tacrine-based multi-target directed Ligands: Enhancing cholinesterase inhibition, NMDA receptor antagonism, and CNS bioavailability for Alzheimer's disease treatment. Eur J Med Chem 2025; 292:117678. [PMID: 40288120 DOI: 10.1016/j.ejmech.2025.117678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Alzheimer's disease (AD) is a multifaceted neurodegenerative disorder for which current treatments provide only symptomatic relief, primarily through cholinesterase (ChE) inhibition and N-methyl-d-aspartate receptor (NMDAR) antagonism. To improve therapeutic efficacy and safety, we designed and synthesized 16 novel tacrine derivatives modified at position 7 with various (hetero)aryl groups or deuterium substitution. Initially, in silico screening predicted favorable CNS permeability and oral bioavailability. Subsequent in vitro evaluations demonstrated significant inhibitory potency against acetylcholinesterase (AChE) and butyrylcholinesterase (BChE), with derivatives 5i and 5m displaying particularly promising profiles. Metabolic stability assessed using human liver microsomes revealed enhanced stability for compound 5e, whereas 5i and 5m underwent rapid metabolism. Notably, compound 7 showed improved metabolic stability attributed to deuterium incorporation. The newly synthesized compounds were further tested for antagonistic activity on the GluN1/GluN2B subtype of NMDAR, with compound 5m exhibiting the most potent and voltage-independent inhibition. The ability of these compounds to permeate the blood-brain barrier (BBB) was confirmed through in vitro PAMPA assays. In preliminary hepatotoxicity screening (HepG2 cells), most derivatives exhibited higher cytotoxicity than tacrine, emphasizing the ongoing challenge in hepatotoxicity management. Based on its overall favorable profile, compound 5m advanced to in vivo pharmacokinetic studies in mice, demonstrating efficient CNS penetration, with brain concentrations exceeding plasma levels (brain-to-plasma ratio 2.36), indicating active transport across the BBB. These findings highlight compound 5m as a promising tacrine-based multi-target-directed ligand, supporting further preclinical development as a potential therapeutic candidate for AD.
Collapse
Affiliation(s)
- Barbora Svobodova
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Zuzana Moravcova
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Anna Misiachna
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Albertov 6, Prague 2, 12843, Czech Republic
| | - Gabriela Novakova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, Prague 6, 166 10, Czech Republic
| | - Ales Marek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, Prague 6, 166 10, Czech Republic
| | - Vladimir Finger
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Jitka Odvarkova
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jana Zdarova Karasova
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Jakub Netolicky
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Albertov 6, Prague 2, 12843, Czech Republic
| | - Marek Ladislav
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Albertov 6, Prague 2, 12843, Czech Republic
| | - Martina Hrabinova
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Ales Sorf
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic; Department of Social and Clinical Pharmacy, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Lubica Muckova
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Lenka Fikejzlova
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic
| | - Marketa Benkova
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Martin Novak
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Lukas Prchal
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic
| | - Jan Capek
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10, Pardubice, Czech Republic
| | - Jiri Handl
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10, Pardubice, Czech Republic
| | - Tomas Rousar
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 532 10, Pardubice, Czech Republic
| | - Katarzyna Ewa Greber
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdansk, Aleja Generała Jozefa Hallera 107, 80-416, Gdansk, Poland
| | - Krzesimir Ciura
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdansk, Aleja Generała Jozefa Hallera 107, 80-416, Gdansk, Poland; Laboratory of Environmental Chemoinformatics, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdansk, Poland
| | - Martin Horak
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic.
| | - Jan Korabecny
- Biomedical Research Center, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic.
| |
Collapse
|
3
|
Kaviyarasu S, Padmanaban N, Khute S, Zengin G, Subash P. Virtual screening and molecular dynamics of anti-Alzheimer compounds from Cardiospermum halicacabum via GC-MS. Front Chem 2025; 13:1586728. [PMID: 40255640 PMCID: PMC12006154 DOI: 10.3389/fchem.2025.1586728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 03/25/2025] [Indexed: 04/22/2025] Open
Abstract
Background Ayurveda is an ancient Indian medicinal system that uses medicinal plants for their neuroprotective effects. Ayurveda claims that the (C. halicacabum) leaves possess significant neuroprotective properties. Alzheimer's is characterized by the accumulation of amyloid-β, acetylcholinesterase, and tau tangles that interfere with neural transmission and impair cognitive abilities. Objectives This study aimed to identify novel potential anti-Alzheimer phytoconstituents of C. halicacabum leaves using in silico methods. Methods This study utilized the Box-Behnken design within the response surface methodology (RSM) to optimize and combine the effects of process variables, namely powder weight, solvent volume, and extraction time, on the microwave-assisted extraction (MAE) of C. halicacabum leaves. The optimization process revealed that these variables, along with microwave usage, significantly influenced the extraction yield. The ethanolic extract was examined using gas chromatography-mass spectrometry (GC-MS) analysis, and the identified phytoconstituents were further analyzed through computer-based simulations, including docking, absorption, distribution, metabolism, excretion, and toxicity (ADMET) studies, assessment of drug-likeness, molecular dynamics, LigPlot analysis, and density functional theory (DFT) analysis. Results Gas chromatography-mass spectrometry (GC-MS) analysis identified 40 phytoconstituents and 37 were successfully characterized. Molecular docking and dynamics simulations revealed two lead compounds, acetic acid (dodecahydro-7-hydroxy-1,4b,8,8-tetramethyl-10-oxo-2(1H)-phenanthrenylidene)-,2-(dimethylamino)ethyl ester, [1R-(1. alpha)], and 1-(2-hydroxyethoxy)-2-methyldodecane, which exhibited superior stability in the docked complex compared to galantamine. Conclusion Based on computational predictions and observed pharmacological properties, these findings suggest that phytoconstituents may have therapeutic effects against selected AD targets.
Collapse
Affiliation(s)
- Selvan Kaviyarasu
- Department of Pharmacognosy, Sri Shanmugha College of Pharmacy, Sankari, Tamil Nadu, India
| | - Nallamuthu Padmanaban
- Department of Pharmacognosy, Sri Shanmugha College of Pharmacy, Sankari, Tamil Nadu, India
| | - Sulekha Khute
- Department of Pharmacognosy, Sri Shanmugha College of Pharmacy, Sankari, Tamil Nadu, India
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Türkiye
| | - Paranthaman Subash
- Department of Pharmacognosy, Sri Shanmugha College of Pharmacy, Sankari, Tamil Nadu, India
| |
Collapse
|
4
|
Cai J, Liu Y, Fan H. Review on pathogenesis and treatment of Alzheimer's disease. Dev Dyn 2025; 254:296-309. [PMID: 39651698 DOI: 10.1002/dvdy.762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 12/11/2024] Open
Abstract
The rising incidence of Alzheimer's disease (AD) and the associated economic impacts has prompted a global focus in the field. In recent years, there has been a growing understanding of the pathogenic mechanisms of AD, including the aggregation of β-amyloid, hyperphosphorylated tau, and neuroinflammation. These processes collectively lead to neurodegeneration and cognitive decline, which ultimately results in the loss of autonomy in patients. Currently, there are three main types of AD treatments: clinical tools, pharmacological treatment, and material interventions. This review provides a comprehensive analysis of the underlying etiology and pathogenesis of AD, as well as an overview of the current prevalence of AD treatments. We believe this article can help deepen our understanding of the AD mechanism, and facilitate the clinical translation of scientific research or therapies, to address this global problem of AD.
Collapse
Affiliation(s)
- Jinxia Cai
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Yanqing Liu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou, China
| |
Collapse
|
5
|
Dehghani S, Ocakcı O, Hatipoglu PT, Özalp VC, Tevlek A. Exosomes as Biomarkers and Therapeutic Agents in Neurodegenerative Diseases: Current Insights and Future Directions. Mol Neurobiol 2025:10.1007/s12035-025-04825-5. [PMID: 40095345 DOI: 10.1007/s12035-025-04825-5] [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: 12/02/2024] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Neurodegenerative diseases (NDs) like Alzheimer's, Parkinson's, and ALS rank among the most challenging global health issues, marked by substantial obstacles in early diagnosis and effective treatment. Current diagnostic techniques frequently demonstrate inadequate sensitivity and specificity, whilst conventional treatment strategies encounter challenges related to restricted bioavailability and insufficient blood-brain barrier (BBB) permeability. Recently, exosomes-nanoscale vesicles packed with proteins, RNAs, and lipids-have emerged as promising agents with the potential to reshape diagnostic and therapeutic approaches to these diseases. Unlike conventional drug carriers, they naturally traverse the BBB and can deliver bioactive molecules to affected neural cells. Their molecular cargo can influence cell signaling, reduce neuroinflammation, and potentially slow neurodegenerative progression. Moreover, exosomes serve as non-invasive biomarkers, enabling early and precise diagnosis while allowing real-time disease monitoring. Additionally, engineered exosomes, loaded with therapeutic molecules, enhance this capability by targeting diseased neurons and overcoming conventional treatment barriers. By offering enhanced specificity, reduced immunogenicity, and an ability to bypass physiological limitations, exosome-based strategies present a transformative advantage over existing diagnostic and therapeutic approaches. This review examines the multifaceted role of exosomes in NDDs, emphasizing their diagnostic capabilities, intrinsic therapeutic functions, and transformative potential as advanced treatment vehicles.
Collapse
Affiliation(s)
- Sam Dehghani
- Faculty of Medicine, Undergraduate Program, Atılım University, 06830, Ankara, Turkey
| | - Ozgecan Ocakcı
- Department of Medical Biology, Faculty of Medicine, AtıLıM University, 06830, Ankara, Turkey
| | - Pars Tan Hatipoglu
- Faculty of Medicine, Undergraduate Program, Atılım University, 06830, Ankara, Turkey
| | - Veli Cengiz Özalp
- Department of Medical Biology, Faculty of Medicine, AtıLıM University, 06830, Ankara, Turkey
| | - Atakan Tevlek
- Department of Medical Biology, Faculty of Medicine, AtıLıM University, 06830, Ankara, Turkey.
| |
Collapse
|
6
|
Gunawardana PBW, Gohil K, Moon KM, Foster LJ, Williams FJ. Proteomic Investigation of Neurotrophic trans-Banglene Reveals Potential Link to Iron Homeostasis. Mol Neurobiol 2025:10.1007/s12035-025-04772-1. [PMID: 40085355 DOI: 10.1007/s12035-025-04772-1] [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: 09/08/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025]
Abstract
In an effort to gain insight into cellular systems impacted by neurotrophic trans-banglene (t-BG), global proteomic profiling and Western blot analyses were employed. Expression level changes in response to t-BG treatment were compared to those observed with nerve growth factor (NGF), a natural neurotrophic protein and functional analog to t-BG. Findings from these studies did not point to direct interception of NGF/TrkA signaling by t-BG. Instead, significant alterations in iron-binding and iron-regulating proteins were observed. While total iron levels showed no change across all treatments, intracellular iron measurements and mitochondrial iron measurements demonstrated lower ferrous (Fe2+) ion levels in t-BG treated cells but not in NGF treated cells. These results highlight a potential connection between iron regulation and neurotrophic activity, a relationship which has, to date, not been well studied. These results are also notable given that iron dysregulation occurs in most neurodegenerative disease settings, and that iron has been shown to facilitate protein aggregation and apoptotic mechanisms.
Collapse
Affiliation(s)
| | - Khyati Gohil
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Kyung-Mee Moon
- Department of Biochemistry & Molecular Biology, University of British Colombia, Vancouver, Canada
| | - Leonard J Foster
- Department of Biochemistry & Molecular Biology, University of British Colombia, Vancouver, Canada
| | | |
Collapse
|
7
|
Zelek WM, Bevan RJ, Nimmo J, Dewilde M, De Strooper B, Morgan BP. Brain-penetrant complement inhibition mitigates neurodegeneration in an Alzheimer's disease mouse model. Brain 2025; 148:941-954. [PMID: 39215579 PMCID: PMC11884734 DOI: 10.1093/brain/awae278] [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: 04/03/2024] [Revised: 07/10/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Complement activation is implicated in driving brain inflammation, self-cell damage and progression of injury in Alzheimer's disease and other neurodegenerative diseases. Here, we investigate the impact of brain delivery of a complement-blocking antibody on neurodegeneration in an Alzheimer's mouse model. We engineered a brain-penetrant recombinant antibody targeting the pro-inflammatory membrane attack complex. Systemic administration of this antibody in APPNL-G-F mice reduced brain levels of complement activation products, demonstrating successful brain entry and target engagement. Prolonged treatment decreased synapse loss, amyloid burden and brain inflammatory cytokine levels, concomitant with cognitive improvement compared to controls. These results underscore the potential of brain-penetrant complement-inhibiting drugs as promising therapeutics, targeting downstream of amyloid plaques in Alzheimer's disease.
Collapse
Affiliation(s)
- Wioleta M Zelek
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| | - Ryan J Bevan
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| | - Jacqui Nimmo
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| | - Maarten Dewilde
- Therapeutic and Diagnostic Antibodies, Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven 3000, Belgium
- PharmAbs, The KU Leuven Antibody Center, KU Leuven, Leuven 3000, Belgium
| | - Bart De Strooper
- Centre for Brain and Disease Research, KU Leuven and VIB Leuven, Leuven 3000, Belgium
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Bryan Paul Morgan
- School of Medicine, UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
8
|
Hatakawa Y, Tanaka A, Furubayashi T, Katsumi H, Nakamura R, Konishi M, Akizawa T, Sakane T. Efficient nose-to-brain delivery of nine residues peptide (JAL-TA9) exhibiting hydrolytic activity against amyloid-β. Eur J Pharm Biopharm 2025; 208:114661. [PMID: 39914573 DOI: 10.1016/j.ejpb.2025.114661] [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: 10/17/2024] [Revised: 12/26/2024] [Accepted: 02/03/2025] [Indexed: 02/23/2025]
Abstract
JAL-TA9 (YKGSGFRMI), is a 9-residue catalytic peptide that cleaves amyloid β (Aβ) 42. Nasal administration was chosen to bypass the blood-brain barrier for efficient brain delivery of JAL-TA9 to treat Alzheimer's disease (AD). Plasma clearance of JAL-TA9 after intravenous bolus injection in rats was very rapid, with a half-life of <1 min. The stability of JAL-TA9 in cerebrospinal fluid (CSF) was much better than that in plasma and whole blood in vitro, suggesting the advantage of direct nasal delivery to avoid rapid elimination from the blood. JAL-TA9 in CSF was 0.115 μg/mL 10 min after nasal administration to rats, but below the detection limit after intravenous injection, indicating a significant direct delivery of JAL-TA9 to the brain. In mice brain, JAL-TA9 concentration was higher after nasal administration than that after intraperitoneal administration. Additionally, peak concentration in the olfactory bulb (OB) after nasal application was at 5 min, whereas in the frontal and occipital brains peaked at 30 and 60 min, respectively, suggesting sequential backward translocation of JAL-TA9 within the brain after direct transport from the nasal cavity to the OB. Therefore, nasal administration allows the efficient delivery of JAL-TA9 to the brain and may be a potential in AD treatment.
Collapse
Affiliation(s)
- Yusuke Hatakawa
- Department of Bio-Analytical Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan.
| | - Akiko Tanaka
- Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-Machi 4-19-1 Higashinada, Kobe, Hyogo 658-8558, Japan.
| | - Tomoyuki Furubayashi
- Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-Machi 4-19-1 Higashinada, Kobe, Hyogo 658-8558, Japan.
| | - Hidemasa Katsumi
- Laboratory of Pharmaceutics, Faculty of Pharmacy, Osaka Ohtani University, Nishikori-kita 3-11-1, Tondabayashi, Osaka 584-8540, Japan.
| | - Rina Nakamura
- O-Force Co., Ltd., 3454, Irino, Kuroshio-Cho, Hata-Gun, Kochi 789-1931, Japan.
| | - Motomi Konishi
- Department of Integrative Pharmaceutical Science, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-Cho, Hirakata, Osaka 573-0101, Japan.
| | - Toshifumi Akizawa
- O-Force Co., Ltd., 3454, Irino, Kuroshio-Cho, Hata-Gun, Kochi 789-1931, Japan; Laboratory of Pharmacology, School of Medicine, Kochi University, Kohasu, Oko-cho, Nankoku, Kochi 783-8505, Japan.
| | - Toshiyasu Sakane
- Department of Pharmaceutical Technology, Kobe Pharmaceutical University, Motoyamakita-Machi 4-19-1 Higashinada, Kobe, Hyogo 658-8558, Japan.
| |
Collapse
|
9
|
Cai Y, Li Y, Wang Y, Xu Y, Chen T, Xue R, Liu Y, Chen W, Yang X, Liu Z, Bao X, Huang Z. Triple-mode sensing platform for acetylcholinesterase activity monitoring and anti-Alzheimer's drug screening based on a highly stable Cu (I) compound. Biosens Bioelectron 2025; 271:117078. [PMID: 39708491 DOI: 10.1016/j.bios.2024.117078] [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: 09/18/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Acetylcholinesterase (AChE) and AChE inhibitors play critical roles in the early diagnosis and treatment of Alzheimer's disease (AD). Herein, a fluorescence/colorimetry/smartphone triple-mode sensing platform was constructed for both AChE activity monitoring and AChE inhibitor screening by exploring a Cu (I) compound, Cu3I (SR)2 (R = CH2CH2NH2), as a fluorescent probe. In comparison of most other fluorescent probes, Cu3I (SR)2 presented exceptional stability against pH, temperature, UV irradiation, redox agents, and metal ions, as well as good recyclability due to its unique chemical structure. We further found the fluorescence emission of Cu3I (SR)2 could be quenched by MnO2 nanosheet (NS) via inner filter effect, and restored by thiocholine (TCh) generated from the hydrolysis of acetylthiocholine iodide (ATCh) in the catalysis of AChE. On this basis, a fluorescence "turn-on" assay was developed for monitoring AChE activity with a detection limit of 0.03 U/L and a detection range of 0.25-50 U/L. This method demonstrates great potential for real-time detection of AChE activity in biological samples and screening of AChE inhibitors obtained from herbal extracts as anti-AD agents. Additionally, Cu3I (SR)2/MnO2 NS sensing system also exhibited a color change from brown to colorless as the increasing AChE activity, which allowed the colorimetric and smartphone detection of AChE activity.
Collapse
Affiliation(s)
- Yanting Cai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 130021, Changchun, China
| | - Yue Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 130021, Changchun, China
| | - Yuanyuan Wang
- College of Chemistry, Jilin University, 130012, Changchun, China
| | - Yihan Xu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 130021, Changchun, China
| | - Tianyan Chen
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 130021, Changchun, China
| | - Ruisong Xue
- College of Chemistry, Jilin University, 130012, Changchun, China
| | - Yanmei Liu
- College of Chemistry, Jilin University, 130012, Changchun, China
| | - Wei Chen
- School of Chips, XJTLU Entrepreneur College (Taicang), Xi'an Jiaotong-Liverpool University, 215400, Suzhou, China
| | - Xiurong Yang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China
| | - Zhen Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 130021, Changchun, China
| | - Xingfu Bao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 130021, Changchun, China
| | - Zhenzhen Huang
- College of Chemistry, Jilin University, 130012, Changchun, China.
| |
Collapse
|
10
|
Yang P, Shuai W, Wang X, Hu X, Zhao M, Wang A, Wu Y, Ouyang L, Wang G. Mitophagy in Neurodegenerative Diseases: Mechanisms of Action and the Advances of Drug Discovery. J Med Chem 2025; 68:3970-3994. [PMID: 39908485 DOI: 10.1021/acs.jmedchem.4c01779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Neurodegenerative diseases (NDDs), such as Parkinson's disease (PD) and Alzheimer's disease (AD), are devastating brain diseases and are incurable at the moment. Increasing evidence indicates that NDDs are associated with mitochondrial dysfunction. Mitophagy removes defective or redundant mitochondria to maintain cell homeostasis, whereas deficient mitophagy accelerates the accumulation of damaged mitochondria to mediate the pathologies of NDDs. Therefore, targeting mitophagy has become a valuable therapeutic pathway for the treatment of NDDs. Several mitophagy modulators have been shown to ameliorate neurodegeneration in PD and AD. However, it remains to be further investigated for other NDDs. Here, we describe the mechanism and key signaling pathway of mitophagy and summarize the roles of defective mitophagy on the pathogenesis of NDDs. Further, we underline the development advances of mitophagy modulators for PD and AD therapy, discuss the therapeutic challenges and limitations of the existing modulators, and provide guidelines for mitophagy mechanism exploration and drug design.
Collapse
Affiliation(s)
- Panpan Yang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Xin Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Xiuying Hu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Aoxue Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Yongya Wu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Sharma A, Rudrawar S, Bharate SB, Jadhav HR. Recent advancements in the therapeutic approaches for Alzheimer's disease treatment: current and future perspective. RSC Med Chem 2025; 16:652-693. [PMID: 39790124 PMCID: PMC11707861 DOI: 10.1039/d4md00630e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
Alzheimer's disease (AD) is a complex, incurable neurological condition characterized by cognitive decline, cholinergic neuron reduction, and neuronal loss. Its exact pathology remains uncertain, but multiple treatment hypotheses have emerged. The current treatments, single or combined, alleviate only symptoms and struggle to manage AD due to its multifaceted pathology. The developmental drugs target pivotal disease factors involved in the envisaged hypotheses and include targets such as amyloid aggregation, hyperphosphorylated tau proteins, and receptors like cholinergic, adrenergic, etc. Present-day research focuses on multi-target directed ligands (MTDLs), which inhibit multiple factors simultaneously, helping slow the disease's progression. This review attempts to collate the recent information related to proposed hypotheses for AD etiology. It systematically organizes the advances in various therapeutic options for AD, with a particular emphasis on clinical candidates. Also, it is expected to help medicinal chemists design novel AD treatments based on available information, which could be helpful to AD patients.
Collapse
Affiliation(s)
- Amit Sharma
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani Pilani Campus, Vidya Vihar Pilani 333031 RJ India +91 1596 244183 +91 1596 255 506
| | - Santosh Rudrawar
- The Institute for Biomedicine and Glycomics, Griffith University Gold Coast 4222 Australia
- School of Pharmacy and Medical Sciences, Griffith University Gold Coast 4222 Australia
| | - Sandip B Bharate
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine Canal Road Jammu 181110 India
| | - Hemant R Jadhav
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani Pilani Campus, Vidya Vihar Pilani 333031 RJ India +91 1596 244183 +91 1596 255 506
| |
Collapse
|
12
|
Reddi Sree R, Kalyan M, Anand N, Mani S, Gorantla VR, Sakharkar MK, Song BJ, Chidambaram SB. Newer Therapeutic Approaches in Treating Alzheimer's Disease: A Comprehensive Review. ACS OMEGA 2025; 10:5148-5171. [PMID: 39989768 PMCID: PMC11840625 DOI: 10.1021/acsomega.4c05527] [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: 06/13/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 02/25/2025]
Abstract
Alzheimer's disease (AD) is an aging-related irreversible neurodegenerative disease affecting mostly the elderly population. The main pathological features of AD are the extracellular Aβ plaques generated by APP cleavage through the amyloidogenic pathway, the intracellular neurofibrillary tangles (NFT) resulting from the hyperphosphorylated tau proteins, and cholinergic neurodegeneration. However, the actual causes of AD are unknown, but several studies suggest hereditary mutations in PSEN1 and -2, APOE4, APP, and the TAU genes are the major perpetrators. In order to understand the etiology and pathogenesis of AD, various hypotheses are proposed. These include the following hypotheses: amyloid accumulation, tauopathy, inflammation, oxidative stress, mitochondrial dysfunction, glutamate/excitotoxicity, cholinergic deficiency, and gut dysbiosis. Currently approved therapeutic interventions are donepezil, galantamine, and rivastigmine, which are cholinesterase inhibitors (ChEIs), and memantine, which is an N-methyl-d-aspartate (NMDA) antagonist. These treatment strategies focus on only symptomatic management of AD by attenuating symptoms but not regeneration of neurons or clearance of Aβ plaques and hyperphosphorylated Tau. This review focuses on the pathophysiology, novel therapeutic targets, and disease-altering treatments such as α-secretase modulators, active immunotherapy, passive immunotherapy, natural antioxidant products, nanomaterials, antiamyloid therapy, tau aggregation inhibitors, transplantation of fecal microbiota or stem cells, and microtubule stabilizers that are in clinical trials or still under investigation.
Collapse
Affiliation(s)
- Radhakrishna Reddi Sree
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Manjunath Kalyan
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre
for Experimental Pharmacology & Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Nikhilesh Anand
- Department
of Pharmacology, American University of
Antigua College of Medicine, University Park, Jabberwock Beach Road, Coolidge, Antigua, Barbuda
| | - Sangeetha Mani
- Department
of Pharmacognosy, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and
Research, Porur, Chennai 600116, India
| | - Vasavi Rakesh Gorantla
- Department
of Anatomical Sciences, St. George’s University School of Medicine, St. George’s University, Saint George, Grenada
| | - Meena Kishore Sakharkar
- College
of
Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Byoung-Joon Song
- Section
of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry
and Biophysics, National Institute on Alcohol
Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20892, United States
| | - Saravana Babu Chidambaram
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre
for Experimental Pharmacology & Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| |
Collapse
|
13
|
Guo R, Li D, Li F, Ji L, Liu H, Qiao H, Lv Z, Tang Y, Wang D. Effects of whole-head 810 nm near-infrared therapy on cognitive and neuropsychiatric symptoms in Alzheimer's disease: A pilot study. J Alzheimers Dis 2025:13872877251313819. [PMID: 39910867 DOI: 10.1177/13872877251313819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by significant cognitive and behavioral impairments. Near-infrared (NIR) light treatment has shown potential in cognitive improvement in previous studies. However, clinical trials of NIR for AD remain limited. OBJECTIVE This study investigated the safety and effects of whole-head 810 nm NIR therapy in AD patients, including long-term efficacy. METHODS An open-label pilot study on whole-head NIR treatment for AD patients was conducted. Nine AD patients completed 4-month treatment (810 nm, 100 mW/cm², 30 min/session, 6 sessions weekly). Safety and efficacy were evaluated at baseline, months 2 and 4, and 2-month post-treatment. RESULTS After four months of whole-head NIR treatment, mean changes from baseline on the Mini-Mental State Examination were 3.2 (p = 0.02). Mean changes from baseline on the Alzheimer's Disease Assessment Scale-Cognitive were -5.0 (p = 0.05), mean changes from baseline on the Montreal Cognitive Assessment were 1.9 (p = 0.12). Mean changes from baseline on the Neuropsychiatric Inventory were -4.2 (p = 0.47). These benefits were sustained two months at least. With no device-related adverse effects were reported. CONCLUSIONS Whole-head 810 nm NIR light is safe and offers promising benefits for AD patients. To fully confirm its efficacy, durability, and underlying mechanisms, further large-scale randomized controlled trials are necessary.
Collapse
Affiliation(s)
- Rong Guo
- School of Biological Science and Medical Engineering, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Deyu Li
- School of Biological Science and Medical Engineering, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
- State Key Laboratory of Software Development Environment, State Key Laboratory of Virtual Reality Technology and System, Beihang University, Beijing, China
| | - Fang Li
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, China
| | - Linna Ji
- Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing, China
| | - Hongying Liu
- Rehabilitation Hospital Affiliated to National Rehabilitation Assistive Devices Research Center, Beijing, China
| | - Huiting Qiao
- School of Biological Science and Medical Engineering, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
| | - Zeping Lv
- Rehabilitation Hospital Affiliated to National Rehabilitation Assistive Devices Research Center, Beijing, China
- Key Laboratory of Assistive Technology for Rehabilitation of Elderly Dysfunction, Beijing, China
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Daifa Wang
- School of Biological Science and Medical Engineering, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, China
- Jiangsu Danyang Huichuang Medical Equipment Co., Ltd, Jiangsu, China
| |
Collapse
|
14
|
Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, Chaudhary K, Sharma S, Sinha A, Abomughaid MM, Imran M, Lakhanpal S, Babalghith AO, Abu-Seer EA, Avinash D, Alzahrani HA, Alhindi AA, Iqbal D, Kumar S, Jha NK, Alghamdi S. Inflammatory signaling pathways in Alzheimer's disease: Mechanistic insights and possible therapeutic interventions. Ageing Res Rev 2025; 104:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [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: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The complex pathophysiology of Alzheimer's disease (AD) poses challenges for the development of therapies. Recently, neuroinflammation has been identified as a key pathogenic mechanism underlying AD, while inflammation has emerged as a possible target for the management and prevention of AD. Several prior studies have demonstrated that medications modulating neuroinflammation might lessen AD symptoms, mostly by controlling neuroinflammatory signaling pathways such as the NF-κB, MAPK, NLRP3, etc, and their respective signaling cascade. Moreover, targeting these inflammatory modalities with inhibitors, natural products, and metabolites has been the subject of intensive research because of their anti-inflammatory characteristics, with many studies demonstrating noteworthy pharmacological capabilities and potential clinical applications. Therefore, targeting inflammation is considered a promising strategy for treating AD. This review comprehensively elucidates the neuroinflammatory mechanisms underlying AD progression and the beneficial effects of inhibitors, natural products, and metabolites in AD treatment.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ashish Kumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Swati Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Eman Adnan Abu-Seer
- Department of Epidemiology and Medical Statistic, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, Saudi Arabia
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Hassan A Alzahrani
- Department of Respiratory Care, Medical Cities at the Minister of Interior, MCMOl, Riyadh, Saudi Arabia
| | | | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India; DST-FIST Laboratory, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology (SBT), Galgotias University, Greater Noida, India; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
15
|
Kaštelan S, Gverović Antunica A, Puzović V, Didović Pavičić A, Čanović S, Kovačević P, Vučemilović PAF, Konjevoda S. Non-Invasive Retinal Biomarkers for Early Diagnosis of Alzheimer's Disease. Biomedicines 2025; 13:283. [PMID: 40002697 PMCID: PMC11852429 DOI: 10.3390/biomedicines13020283] [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: 12/12/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the brain associated with ageing and is the most prevalent form of dementia, affecting an estimated 55 million people worldwide, with projections suggesting this number will exceed 150 million by 2050. With its increasing prevalence, AD represents a significant global health challenge with potentially serious social and economic consequences. Diagnosing AD is particularly challenging as it requires timely recognition. Currently, there is no effective therapy for AD; however, certain medications may help slow its progression. Existing diagnostic methods such as magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET), and biomarker analysis in cerebrospinal fluid tend to be expensive and invasive, making them impractical for widespread use. Consequently, research into non-invasive biomarkers that enable early detection and screening for AD is a crucial area of contemporary clinical investigation. One promising approach for the early diagnosis of AD may be retinal imaging. As an extension of the central nervous system, the retina offers a distinctive opportunity for non-invasive brain structure and function assessment. Considering their shared embryological origins and the vascular and immunological similarities between the eye and brain, alterations in the retina may indicate pathological changes in the brain, including those specifically related to AD. Studies suggest that structural and vascular changes in the retina, particularly within the neuronal network and blood vessels, may act as markers of cerebral changes caused by AD. These retinal alterations have the potential to act as biomarkers for early diagnosis. Since AD is typically diagnosed only after a significant neuronal loss has occurred, identifying early diagnostic markers could enable timely intervention and help prevent disease progression. Non-invasive retinal imaging techniques, such as optical coherence tomography (OCT) and OCT angiography, provide accessible methods for the early detection of changes linked to AD. This review article focuses on the potential of retinal imaging as a non-invasive biomarker for early diagnosis of AD. Investigating the ageing of the retina and its connections to neurodegenerative processes could significantly enhance the diagnosis, monitoring, and treatment of AD, paving the way for new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Snježana Kaštelan
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Ophthalmology, Clinical Hospital Dubrava, 10000 Zagreb, Croatia
| | | | - Velibor Puzović
- Department of Pathology, General Hospital Dubrovnik, 20000 Dubrovnik, Croatia
| | | | - Samir Čanović
- Department of Ophthalmology, Zadar General Hospital, 23000 Zadar, Croatia
- Department of Health Studies, University of Zadar, 23000 Zadar, Croatia
| | - Petra Kovačević
- Department of Ophthalmology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | | | - Suzana Konjevoda
- Department of Ophthalmology, Zadar General Hospital, 23000 Zadar, Croatia
- Department of Health Studies, University of Zadar, 23000 Zadar, Croatia
| |
Collapse
|
16
|
Chunhui G, Yanqiu Y, Jibing C, Ning L, Fujun L. Exosomes and non-coding RNAs: bridging the gap in Alzheimer's pathogenesis and therapeutics. Metab Brain Dis 2025; 40:84. [PMID: 39754674 PMCID: PMC11700052 DOI: 10.1007/s11011-024-01520-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that primarily affects the elderly population and is the leading cause of dementia. Meanwhile, the vascular hypothesis suggests that vascular damage occurs in the early stages of the disease, leading to neurodegeneration and hindered waste clearance, which in turn triggers a series of events including the accumulation of amyloid plaques and Tau protein tangles. Non-coding RNAs (ncRNAs), including long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), have been found to be involved in the regulation of AD. Furthermore, lncRNAs and circRNAs can act as competitive endogenous RNAs to inhibit miRNAs, and their interactions can form a complex regulatory network. Exosomes, which are extracellular vesicles (EVs), are believed to be able to transfer ncRNAs between cells, thus playing a regulatory role in the brain by crossing the blood-brain barrier (BBB). Exosomes are part of the intercellular carrier system; therefore, utilizing exosomes to deliver drugs to recipient cells might not activate the immune system, making it a potential strategy to treat central nervous system diseases. In this review, we review that AD is a multifactorial neurological disease and that ncRNAs can regulate its multiple pathogenic mechanisms to improve our understanding of the etiology of AD and to simultaneously regulate multiple pathogenic mechanisms of AD through the binding of ncRNAs to exosomes to improve the treatment of AD.
Collapse
Affiliation(s)
- Guo Chunhui
- Graduate School, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - You Yanqiu
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Chen Jibing
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China.
| | - Luo Ning
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China.
| | - Li Fujun
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China.
| |
Collapse
|
17
|
Shaaban AE, Ali AR, Ayyad SN, Badria FA. Multi-target directed ligands inspired natural products as an effective approach for the treatment of complex chronic health disorders. Bioorg Chem 2025; 154:108075. [PMID: 39708551 DOI: 10.1016/j.bioorg.2024.108075] [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: 10/01/2024] [Revised: 11/30/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Complex diseases involve multifaceted etiological components, which limit the effectiveness of conventional targeted therapies. Therefore, standard medicinal treatments often face significant challenges and failures when addressing these disease conditions. Furthermore, the growing interest in multidrug resistance (MDR), the occurrence of adverse drug reactions related to use traditional approaches, and the limited clinical efficacy of single-target drug therapy have increased the demand for innovative drug treatments. In this rapidly evolving era, the exploration of multi-target directed ligands (MTDLs) derived from natural products has granted us access to a wide range of compounds with medicinal properties. The allure of these MTDLs lies in their unique ability to minimize side effects from using two medicinal agents, establishing them as the preferred choice for drug developers. MTDLs have been recognized for their extraordinary capacity to collectively hinder multiple pathways implicated in the development of intricate diseases by merging or linking active molecules obtained from these sources. This review delves into promising MTDLs derived from natural products, which modulates diverse biological pathways implicated in complex diseased conditions particularly Alzheimer's disease, diabetes, cardiac disorders and inflammatory conditions.
Collapse
Affiliation(s)
- Amira E Shaaban
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt
| | - Ahmed R Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Seif N Ayyad
- Department of Organic Chemistry, Faculty of Science, Damietta University, New Damietta 34511, Egypt
| | - Farid A Badria
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
18
|
Satyanarayanan SK, Han Z, Xiao J, Yuan Q, Yung WH, Ke Y, Chang RCC, Zhu MH, Su H, Su KP, Qin D, Lee SMY. Frontiers of Neurodegenerative Disease Treatment: Targeting Immune Cells in Brain Border Regions. Brain Behav Immun 2025; 123:483-499. [PMID: 39378973 DOI: 10.1016/j.bbi.2024.10.007] [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: 05/03/2024] [Revised: 09/15/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024] Open
Abstract
Neurodegenerative diseases (NDs) demonstrate a complex interaction with the immune system, challenging the traditional view of the brain as an "immune-privileged" organ. Microglia were once considered the sole guardians of the brain's immune response. However, recent research has revealed the critical role of peripheral immune cells located in key brain regions like the meninges, choroid plexus, and perivascular spaces. These previously overlooked cells are now recognized as contributors to the development and progression of NDs. This newfound understanding opens doors for pioneering therapeutic strategies. By targeting these peripheral immune cells, we may be able to modulate the brain's immune environment, offering an alternative approach to treat NDs and circumvent the challenges posed by the blood-brain barrier. This comprehensive review will scrutinize the latest findings on the complex interactions between these peripheral immune cells and NDs. It will also critically assess the prospects of targeting these cells as a ground-breaking therapeutic avenue for these debilitating disorders.
Collapse
Affiliation(s)
- Senthil Kumaran Satyanarayanan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Zixu Han
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Jingwei Xiao
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Qiuju Yuan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing Ho Yung
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Ya Ke
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Faculty of Medicine Building, Hong Kong, China
| | - Maria Huachen Zhu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, Hong Kong, China
| | - Kuan-Pin Su
- An-Nan Hospital, China Medical University, Tainan, Taiwan; College of Medicine, China Medical University, Taichung, Taiwan; Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
| | - Dajiang Qin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| | - Suki Man Yan Lee
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510799, China; School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
19
|
Șovrea AS, Boșca AB, Dronca E, Constantin AM, Crintea A, Suflețel R, Ștefan RA, Ștefan PA, Onofrei MM, Tschall C, Crivii CB. Non-Drug and Non-Invasive Therapeutic Options in Alzheimer's Disease. Biomedicines 2025; 13:84. [PMID: 39857667 PMCID: PMC11760896 DOI: 10.3390/biomedicines13010084] [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: 12/16/2024] [Revised: 12/28/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
Despite the massive efforts of modern medicine to stop the evolution of Alzheimer's disease (AD), it affects an increasing number of people, changing individual lives and imposing itself as a burden on families and the health systems. Considering that the vast majority of conventional drug therapies did not lead to the expected results, this review will discuss the newly developing therapies as an alternative in the effort to stop or slow AD. Focused Ultrasound (FUS) and its derived Transcranial Pulse Stimulation (TPS) are non-invasive therapeutic approaches. Singly or as an applied technique to change the permeability of the blood-brain-barrier (BBB), FUS and TPS have demonstrated the benefits of use in treating AD in animal and human studies. Adipose-derived stem Cells (ADSCs), gene therapy, and many other alternative methods (diet, sleep pattern, physical exercise, nanoparticle delivery) are also new potential treatments since multimodal approaches represent the modern trend in this disorder research therapies.
Collapse
Affiliation(s)
- Alina Simona Șovrea
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Adina Bianca Boșca
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Eleonora Dronca
- Molecular Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (E.D.); (A.C.)
| | - Anne-Marie Constantin
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Andreea Crintea
- Molecular Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (E.D.); (A.C.)
| | - Rada Suflețel
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Roxana Adelina Ștefan
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Paul Andrei Ștefan
- Radiology and Imaging Department, Emergency County Hospital Cluj, 400347 Cluj-Napoca, Romania;
| | - Mădălin Mihai Onofrei
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Christoph Tschall
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| | - Carmen-Bianca Crivii
- Morpho-Functional Sciences Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania; (A.S.Ș.); (A.-M.C.); (R.S.); (R.A.Ș.); (M.M.O.); (C.-B.C.)
| |
Collapse
|
20
|
Dash R, Samanta S, Jena BR, Pradhan S. Nanotechnology in Drug Delivery: An Overview of Developing the Blood Brain Barrier. Curr Neurovasc Res 2025; 21:347-358. [PMID: 39411960 DOI: 10.2174/0115672026346307240919112023] [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: 07/27/2023] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 02/27/2025]
Abstract
The close connection between the brain microvascular endothelial cells (BMECs) that are enclosed within this barrier is the result of an intracellular junction, which is responsible for the constricted connection. The regulation and control of drug delivery systems both require nanoparticles, which are extremely small particles made up of a variety of materials, including polymers, metals, and other chemicals. Nanoparticles are a crucial component of the regulation and control of drug delivery systems. There is a possibility that nanomaterials composed of inorganic chemicals, such as gold nanoparticles, could be utilized in the treatment of neurodegenerative illnesses like Parkinson's disease. In addition to this, they are used as nano-carriers for the aim of distributing drugs to the region of the brain that is being targeted. There are a number of advantages that are easily apparent when compared to other methods of administering drugs for neurological diseases. The current review demonstrates both the advantages and disadvantages of utilizing a wide variety of nanomaterials for brain delivery, as well as the potential impact that this will have in the future on the safety and effectiveness of patient care.
Collapse
Affiliation(s)
- Rasmita Dash
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Subhankar Samanta
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Bikash Ranjan Jena
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Soumyaranjan Pradhan
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| |
Collapse
|
21
|
Pandey K, Srivastava P, Pandey SK, Johari S, Bhatnagar P, Sonane M, Mishra A. Stem Cells as a Novel Source for Regenerative Medicinal Applications in Alzheimer's Disease: An Update. Curr Mol Med 2025; 25:146-166. [PMID: 39318206 DOI: 10.2174/0115665240334785240913071442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder characterized by loss of the neurons, excessive accumulation of misfolded Aβ and Tau proteins, and degeneration of neural synapses, primarily occurring in the neocortex and the hippocampus regions of the brain. AD Progression is marked by cognitive deterioration, memory decline, disorientation, and loss of problem-solving skills, as well as language. Due to limited comprehension of the factors contributing to AD and its severity due to neuronal loss, even today, the medications approved by the U.S. Food and Drug Administration (FDA) are not precisely efficient and curative. Stem cells possess great potential in aiding AD due to their self-renewal, proliferation, and differentiation properties. Stem cell therapy can aid by replacing the lost neurons, enhancing neurogenesis, and providing an enriched environment to the pre-existing neural cells. Stem cell therapy has provided us with promising results in regard to the animal AD models, and even pre-clinical studies have shown rather positive results. Cell replacement therapies are potential curative means to treat AD, and there are a number of undergoing human clinical trials to make Stem Cell therapy accessible for AD patients. In this review, we aim to discuss the AD pathophysiology and varied stem cell types and their application.
Collapse
Affiliation(s)
- Kratika Pandey
- School of Biosciences, Institute of Management Studies Ghaziabad 9 (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, 201015, India
| | - Priyanka Srivastava
- School of Biosciences, Institute of Management Studies Ghaziabad 9 (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, 201015, India
| | - Swaroop Kumar Pandey
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| | - Surabhi Johari
- School of Biosciences, Institute of Management Studies Ghaziabad 9 (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, 201015, India
| | - Priyanka Bhatnagar
- Department of Pharmacology, All India Institute of Medical Sciences, Ansari Nagar, Delhi, 110029, India
| | - Madhavi Sonane
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, 226025, India
| | - Anuja Mishra
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281406, India
| |
Collapse
|
22
|
Abidar S, Hritcu L, Nhiri M. An Overview of the Natural Neuroprotective Agents for the Management of Cognitive Impairment Induced by Scopolamine in Zebrafish ( Danio rerio). CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:21-31. [PMID: 39039682 DOI: 10.2174/0118715273309256240702053609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder mainly characterized by dementia and cognitive decline. AD is essentially associated with the presence of aggregates of the amyloid-β peptide and the hyperphosphorylated microtubule-associated protein tau. The available AD therapies can only alleviate the symptoms; therefore, the development of natural treatments that exhibit neuroprotective effects and correct the behavioral impairment is a critical requirement. The present review aims to collect the natural substances that have been evaluated for their neuroprotective profile against AD-like behaviors induced in zebrafish (Danio rerio) by scopolamine. We focused on articles retrieved from the PubMed database via preset searching strings from 2010 to 2023. Our review assembled 21 studies that elucidated the activities of 28 various natural substances, including bioactive compounds, extracts, fractions, commercial compounds, and essential oils. The listed compounds enhanced cognition and showed several mechanisms of action, namely antioxidant potential, acetylcholinesterase's inhibition, and reduction of lipid peroxidation. Additional studies should be achieved to demonstrate their preventive and therapeutic activities in cellular and rodent models. Further clinical trials would be extremely solicited to support more insight into the neuroprotective effects of the most promising drugs in an AD context.
Collapse
Affiliation(s)
- Sara Abidar
- Laboratory of Biochemistry and Molecular Genetics (LBMG), Faculty of Sciences and Technologies of Tangier (FSTT) Abdelmalek Essaadi University, Tetouan, Morocco
| | - Lucian Hritcu
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 700506 Iasi, Romania
| | - Mohamed Nhiri
- Laboratory of Biochemistry and Molecular Genetics (LBMG), Faculty of Sciences and Technologies of Tangier (FSTT) Abdelmalek Essaadi University, Tetouan, Morocco
| |
Collapse
|
23
|
Xing C, Chen H, Bi W, Lei T, Hang Z, Du H. Targeting 5-HT Is a Potential Therapeutic Strategy for Neurodegenerative Diseases. Int J Mol Sci 2024; 25:13446. [PMID: 39769209 PMCID: PMC11679250 DOI: 10.3390/ijms252413446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
There is increasing interest in the potential therapeutic role of 5-HT (serotonin) in the treatment of neurodegenerative diseases, which are characterized by the progressive degeneration and death of nerve cells. 5-HT is a vital neurotransmitter that plays a central role in regulating mood, cognition, and various physiological processes in the body. Disruptions in the 5-HT system have been linked to several neurological and psychiatric disorders, making it an attractive target for therapeutic intervention. Although the exact causes of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) are not fully understood, researchers believe that regulating the 5-HT system could help alleviate symptoms and potentially slow the progression of these diseases. Here, we delve into the potential of harnessing 5-HT as a therapeutic target for the treatment of neurodegenerative diseases. It is important to note that the current clinical drugs targeting 5-HT are still limited in the treatment of these complex diseases. Therefore, further research and clinical trials are needed to evaluate the feasibility and effectiveness of its clinical application.
Collapse
Affiliation(s)
- Cencan Xing
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Hongyu Chen
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Wangyu Bi
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Tong Lei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Zhongci Hang
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Hongwu Du
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| |
Collapse
|
24
|
Minuti A, Mazzon E, Iori R, Chiricosta L, Artimagnella O. Bioactivated Glucoraphanin Improves Cell Survival, Upregulating Phospho-AKT, and Modulates Genes Involved in DNA Repair in an In Vitro Alzheimer's Disease Model: A Network-Transcriptomic Analysis. Nutrients 2024; 16:4202. [PMID: 39683594 DOI: 10.3390/nu16234202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Alzheimer's disease (AD) is one of the most common neurodegenerative diseases, for which a definitive cure is still missing. Recently, natural compounds have been investigated for their possible neuroprotective role, including the bioactivated product of glucoraphanin (GRA), the sulforaphane (SFN), which is highly rich in cruciferous vegetables. It is known that SFN alleviates neuronal dysfunction, apoptosis, and oxidative stress in the brain. In the light of this evidence, the aim of this study was to investigate the molecular effects of SFN pre-treatment in differentiated SH-SY5Y neurons exposed to β-amyloid (Aβ). METHODS To this end, we first evaluated first cell viability via the Thiazolyl Blue Tetrazolium Bromide (MTT) assay, and then we analyzed the transcriptomic profiles by next-generation sequencing (NGS). Finally, we used a network analysis in order to understand which biological processes are affected, validating them by Western blot assay. RESULTS SFN pre-treatment counteracted Aβ-induced loss of cell viability. The network-transcriptomic analysis revealed that SFN upregulates genes associated with DNA repair, such as ABRAXAS1, BRCA1, BRCA2, CDKN1A, FANCA, FANCD2, FANCE, NBN, and XPC. Finally, SFN also increased the phosphorylation of AKT, which is associated with DNA repair and cell survival. CONCLUSIONS These data suggest that SFN is a natural compound that could be suitable in the prevention of AD, thanks to its neuroprotective role in increasing cell survival, potentially restoring DNA damage induced by Aβ exposure.
Collapse
Affiliation(s)
- Aurelio Minuti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Emanuela Mazzon
- Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio" Chieti-Pescara, 66100 Chieti, Italy
| | - Renato Iori
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach (FEM), Via E. Mach 1, 38098 San Michele all'Adige, Italy
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Osvaldo Artimagnella
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
25
|
Célestine M, Jacquier-Sarlin M, Borel E, Petit F, Lante F, Bousset L, Hérard AS, Buisson A, Dhenain M. Transmissible long-term neuroprotective and pro-cognitive effects of 1-42 beta-amyloid with A2T icelandic mutation in an Alzheimer's disease mouse model. Mol Psychiatry 2024; 29:3707-3721. [PMID: 38871852 PMCID: PMC11609088 DOI: 10.1038/s41380-024-02611-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024]
Abstract
The amyloid cascade hypothesis assumes that the development of Alzheimer's disease (AD) is driven by a self-perpetuating cycle, in which β-amyloid (Aβ) accumulation leads to Tau pathology and neuronal damages. A particular mutation (A673T) of the amyloid precursor protein (APP) was identified among Icelandic population. It provides a protective effect against Alzheimer- and age-related cognitive decline. This APP mutation leads to the reduced production of Aβ with A2T (position in peptide sequence) change (Aβice). In addition, Aβice has the capacity to form protective heterodimers in association with wild-type Aβ. Despite the emerging interest in Aβice during the last decade, the impact of Aβice on events associated with the amyloid cascade has never been reported. First, the effects of Aβice were evaluated in vitro by electrophysiology on hippocampal slices and by studying synapse morphology in cortical neurons. We showed that Aβice protects against endogenous Aβ-mediated synaptotoxicity. Second, as several studies have outlined that a single intracerebral administration of Aβ can worsen Aβ deposition and cognitive functions several months after the inoculation, we evaluated in vivo the long-term effects of a single inoculation of Aβice or Aβ-wild-type (Aβwt) in the hippocampus of transgenic mice (APPswe/PS1dE9) over-expressing Aβ1-42 peptide. Interestingly, we found that the single intra-hippocampal inoculation of Aβice to mice rescued synaptic density and spatial memory losses four months post-inoculation, compared with Aβwt inoculation. Although Aβ load was not modulated by Aβice infusion, the amount of Tau-positive neuritic plaques was significantly reduced. Finally, a lower phagocytosis by microglia of post-synaptic compounds was detected in Aβice-inoculated animals, which can partly explain the increased density of synapses in the Aβice animals. Thus, a single event as Aβice inoculation can improve the fate of AD-associated pathology and phenotype in mice several months after the event. These results open unexpected fields to develop innovative therapeutic strategies against AD.
Collapse
Affiliation(s)
- Marina Célestine
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, MIRCen, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France
| | - Muriel Jacquier-Sarlin
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, GIN, 38000, Grenoble, France
| | - Eve Borel
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, GIN, 38000, Grenoble, France
| | - Fanny Petit
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, MIRCen, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France
| | - Fabien Lante
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, GIN, 38000, Grenoble, France
| | - Luc Bousset
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, MIRCen, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France
| | - Anne-Sophie Hérard
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, MIRCen, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France
| | - Alain Buisson
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, GIN, 38000, Grenoble, France
| | - Marc Dhenain
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France.
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Direction de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, MIRCen, 18 Route du Panorama, F-92265, Fontenay-aux-Roses, France.
| |
Collapse
|
26
|
Merighi S, Nigro M, Travagli A, Fernandez M, Vincenzi F, Varani K, Pasquini S, Borea PA, Salati S, Cadossi R, Gessi S. Effect of Low-Frequency, Low-Energy Pulsed Electromagnetic Fields in Neuronal and Microglial Cells Injured with Amyloid-Beta. Int J Mol Sci 2024; 25:12847. [PMID: 39684558 DOI: 10.3390/ijms252312847] [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: 10/09/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative pathology covering about 70% of all cases of dementia. It is associated with neuroinflammation and neuronal cell death, which are involved in disease progression. There is a lack of effective therapies, and halting this process represents a therapeutic challenge. Data in the literature suggest several neuroprotective effects of low-frequency, low-energy pulsed electromagnetic fields (PEMFs) on biological systems, and clinical studies report that PEMF stimulation is safe and well tolerated. The aim of this work is to investigate the effects of PEMF exposure on oxidative stress and cell death in in vitro-injured cellular models of neurons and microglia. SH-SY5Y cells were stimulated by hydrogen peroxide (H2O2) or amyloid-β (Aβ) peptide, and N9 microglial cells were activated with lipopolysaccharide (LPS) or Aβ peptide. Reactive oxygen production, mitochondrial integrity, and cell death modulation were investigated through 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) and 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolocarbo-cyanine iodide (JC-1) biochemical assays, fluorescence, and MTS experiments. Cells were exposed to PEMFs producing a pulsed signal with the following parameters: pulse duration of 1.3 ms and frequency of 75 Hz. The outcomes demonstrated that PEMFs defended SH-SY5Y cells against Aβ peptide- or H2O2-induced oxidative stress, mitochondrial damage, and cell death. Furthermore, in microglia activated by LPS or Aβ peptide, they reverted the reduction in mitochondrial potential, oxidative damage, and cell death. Overall, these findings imply that PEMFs influence the redox state of the cells by significantly boosting antioxidant levels in both injured microglia and neuronal in vitro cells mimicking in vitro AD.
Collapse
Affiliation(s)
- Stefania Merighi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Manuela Nigro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Alessia Travagli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Mercedes Fernandez
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Silvia Pasquini
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy
| | | | - Simona Salati
- Igea Clinical Biophysics, Medical Division, 41012 Carpi, Italy
| | - Ruggero Cadossi
- Igea Clinical Biophysics, Medical Division, 41012 Carpi, Italy
| | - Stefania Gessi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
27
|
Qi X, Nizamutdinov D, Yi SS, Wu E, Huang JH. Disease Modifying Monoclonal Antibodies and Symptomatic Pharmacological Treatment for Alzheimer's Disease. Biomedicines 2024; 12:2636. [PMID: 39595200 PMCID: PMC11592475 DOI: 10.3390/biomedicines12112636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's Disease (AD) is an irreversible, progressive syndrome characterized by neurocognitive impairment. Two neuropathological features seen in AD are extracellular amyloid plaques consisting of amyloid beta1-40 and 1-42, and intracellular neurofibrillary tangles (NFTs). For decades, neuroscience research has heavily focused on seeking to understand the primary mechanism of AD and searching for pharmacological approaches for the treatment of dementia. Three monoclonal antibodies that act against amyloid beta-aducanumab, lecanemab, and donanemab-have been approved by the Food and Drug Administration (FDA) for the treatment of mild cognitive impairment and mild AD, in addition to medications for cognitive symptom management such as acetylcholinesterase inhibitors and the N-methyl-D-aspartate (NMDA) antagonist. Further trials should focus on the combination of therapies targeting amyloid plaques and tau pathology.
Collapse
Affiliation(s)
- Xiaoming Qi
- Department of Neurology, Baylor Scott & White Health, Temple, TX 78508, USA;
| | - Damir Nizamutdinov
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
| | - Song Stephen Yi
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
| | - Erxi Wu
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX 76508, USA
- College of Medicine, Texas A&M University, College Station, TX 77843, USA
- College of Irma Lerma Rangel College of Pharmacy, Texas A&M University, College Station, TX 77843, USA
- LIVESTRONG Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jason H. Huang
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott & White Health, Temple, TX 78508, USA; (D.N.); (S.S.Y.); (E.W.)
- Department of Neurosurgery, Baylor College of Medicine, Temple, TX 76508, USA
- College of Medicine, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
28
|
Mishra KA, Sethi KK. Unveiling tomorrow: Carbonic anhydrase activators and inhibitors pioneering new frontiers in Alzheimer's disease. Arch Pharm (Weinheim) 2024:e2400748. [PMID: 39506506 DOI: 10.1002/ardp.202400748] [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: 09/16/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and a principal basis of dementia in the elderly population globally. Recently, human carbonic anhydrases (hCAs, EC 4.2.1.1) were demonstrated as possible new targets for treating AD. hCAs are vital for maintaining pH balance and performing other physiological processes as they catalyze the reversible hydration of carbon dioxide to bicarbonate and a proton. Current research indicates that hCA plays a role in brain functions critical for transmitting neural signals. Activation of carbonic anhydrase (CA) has emerged as a promising avenue in addressing memory loss and cognitive issues. Conversely, the exploration of CA inhibition represents a novel frontier in this field. By enhancing glial fitness and cerebrovascular health and blocking amyloid-β (Aβ)-induced mitochondrial dysfunction pathways, cytochrome C (CytC) release, caspase 9 activation, and H2O2 generation in neurons, CA inhibitors improve cognition and lessen the pathology caused by Aβ. Recent research has pushed hCAs into the spotlight as critical players in AD pathogenesis and precise therapeutic targets. The captivating dilemma of choosing between hCA inhibitors and activators looms large, as inhibitors reduce Aβ aggregation and improve cerebral blood flow, while activators enhance cerebrovascular functions and restore pH balance. The current review sheds light on the clinical evidence for hCAs and the roles of inhibitors and activators in AD. Additionally, this review offers a fascinating outlook on the data that may aid medicinal chemists in designing and developing new leads that are more effective and selective for upcoming in vitro and in vivo studies, allowing for the discovery and introduction of novel drug candidates for the treatment of AD to the market and into the clinical pipeline.
Collapse
Affiliation(s)
- Km Abha Mishra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Guwahati, Assam, India
| | - Kalyan K Sethi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Guwahati, Guwahati, Assam, India
| |
Collapse
|
29
|
Thawabteh AM, Ghanem AW, AbuMadi S, Thaher D, Jaghama W, Karaman D, Karaman R. Recent Advances in Therapeutics for the Treatment of Alzheimer's Disease. Molecules 2024; 29:5131. [PMID: 39519769 PMCID: PMC11547905 DOI: 10.3390/molecules29215131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The most prevalent chronic neurodegenerative illness in the world is Alzheimer's disease (AD). It results in mental symptoms including behavioral abnormalities and cognitive impairment, which have a substantial financial and psychological impact on the relatives of the patients. The review discusses various pathophysiological mechanisms contributing to AD, including amyloid beta, tau protein, inflammation, and other factors, while emphasizing the need for effective disease-modifying therapeutics that alter disease progression rather than merely alleviating symptoms. This review mainly covers medications that are now being studied in clinical trials or recently approved by the FDA that fall under the disease-modifying treatment (DMT) category, which alters the progression of the disease by targeting underlying biological mechanisms rather than merely alleviating symptoms. DMTs focus on improving patient outcomes by slowing cognitive decline, enhancing neuroprotection, and supporting neurogenesis. Additionally, the review covers amyloid-targeting therapies, tau-targeting therapies, neuroprotective therapies, and others. This evaluation specifically looked at studies on FDA-approved novel DMTs in Phase II or III development that were carried out between 2021 and 2024. A thorough review of the US government database identified clinical trials of biologics and small molecule drugs for 14 agents in Phase I, 34 in Phase II, and 11 in Phase III that might be completed by 2028.
Collapse
Affiliation(s)
- Amin Mahmood Thawabteh
- Department of Chemistry, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine;
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Aseel Wasel Ghanem
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Sara AbuMadi
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Dania Thaher
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Weam Jaghama
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit P.O. Box 14, West Bank, Palestine; (A.W.G.); (S.A.); (D.T.); (W.J.)
| | - Donia Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 20002, Palestine;
| | - Rafik Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 20002, Palestine;
- Department of Sciences, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| |
Collapse
|
30
|
Peng L, Zhang Z, Li Q, Song Z, Yan C, Ling H. Unveiling the multifaceted pathogenesis and therapeutic drugs of Alzheimer's disease: A comprehensive review. Heliyon 2024; 10:e39217. [PMID: 39629139 PMCID: PMC11612466 DOI: 10.1016/j.heliyon.2024.e39217] [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: 03/31/2024] [Revised: 08/02/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder characterized by the accumulation of β-amyloid (Aβ) plaques and tau phosphorylation-induced neurofibrillary tangles. This review comprehensively summarizes AD pathogenesis and related factors, drawing on a wealth of authoritative reports and research findings. Specifically, we delve into the intricate mechanisms underlying AD pathology, including Aβ deposition, tau protein phosphorylation, cholinergic dysfunction, neuroinflammation, mitochondrial oxidative stress, ferroptosis, imbalance in the gut microbiota, and microRNA dysregulation. We also explored the effects of these factors on the brain, including synaptic damage and cognitive impairment. Moreover, our review highlights the associations between the pathogenesis of AD and inflammatory cytokines in the peripheral blood and cerebrospinal fluid, dysbiosis of the gut microbiota, and changes in microRNA expression. Overall, we provided a systematic and illustrative overview of the pathogenesis and therapeutic drugs for AD, offering help in the prevention and treatment of this condition.
Collapse
Affiliation(s)
- Liting Peng
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| | - Zhiming Zhang
- Department of Anesthesiology, The First People's Hospital of Chenzhou, The Chenzhou Affiliated Hospital, Hengyang Medical School, University of South China, Chenzhou, 423000, Hunan, China
| | - Qi Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| | - Zhenjiang Song
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| | - Canqun Yan
- The Health Management Center, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hongyan Ling
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| |
Collapse
|
31
|
Capocchi JK, Figueroa-Romero C, Dunham SJB, Faraci G, Rothman JA, Whiteson KL, Seo DO, Holtzman DM, Grabrucker S, Nolan YM, Kaddurah-Daouk R, Jett DA. Symposium: What Does the Microbiome Tell Us about Prevention and Treatment of AD/ADRD? J Neurosci 2024; 44:e1295242024. [PMID: 39384409 PMCID: PMC11466070 DOI: 10.1523/jneurosci.1295-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 07/17/2024] [Accepted: 08/19/2024] [Indexed: 10/11/2024] Open
Abstract
Alzheimer's disease (AD) and Alzheimer's disease-related dementias (ADRDs) are broad-impact multifactorial neurodegenerative diseases. Their complexity presents unique challenges for developing effective therapies. This review highlights research presented at the 2024 Society for Neuroscience meeting which emphasized the gut microbiome's role in AD pathogenesis by influencing brain function and neurodegeneration through the microbiota-gut-brain axis. This emerging evidence underscores the potential for targeting the gut microbiota to treat AD/ADRD.
Collapse
Affiliation(s)
| | | | | | - Gina Faraci
- University of California, Irvine, Irvine, California 92697
| | - Jason A Rothman
- University of California, Irvine, Irvine, California 92697
- University of California, Riverside, Riverside, California 92521
| | | | - Dong-Oh Seo
- Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - David M Holtzman
- Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - Stefanie Grabrucker
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 XF62, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 XF62, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
| | | | - David A Jett
- National Institute of Neurological Disorders and Stroke, Rockville, Maryland 20852
| |
Collapse
|
32
|
Gonçalves PB, Sodero ACR, Cordeiro Y. Natural products targeting amyloid-β oligomer neurotoxicity in Alzheimer's disease. Eur J Med Chem 2024; 276:116684. [PMID: 39032401 DOI: 10.1016/j.ejmech.2024.116684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Alzheimer's disease (AD) constitutes a major global health issue, characterized by progressive neurodegeneration and cognitive impairment, for which no curative treatment is currently available. Current therapeutic approaches are focused on symptom management, highlighting the critical need for disease-modifying therapy. The hallmark pathology of AD involves the aggregation and accumulation of amyloid-β (Aβ) peptides in the brain. Consequently, drug discovery efforts in recent decades have centered on the Aβ aggregation cascade, which includes the transition of monomeric Aβ peptides into toxic oligomers and, ultimately, mature fibrils. Historically, anti-Aβ strategies focused on the clearance of amyloid fibrils using monoclonal antibodies. However, substantial evidence has highlighted the critical role of Aβ oligomers (AβOs) in AD pathogenesis. Soluble AβOs are now recognized as more toxic than fibrils, directly contributing to synaptic impairment, neuronal damage, and the onset of AD. Targeting AβOs has emerged as a promising therapeutic approach to mitigate cognitive decline in AD. Natural products (NPs) have demonstrated promise against AβO neurotoxicity through various mechanisms, including preventing AβO formation, enhancing clearance mechanisms, or converting AβOs into non-toxic species. Understanding the mechanisms by which anti-AβO NPs operate is useful for developing disease-modifying treatments for AD. In this review, we explore the role of NPs in mitigating AβO neurotoxicity for AD drug discovery, summarizing key evidence from biophysical methods, cellular assays, and animal models. By discussing how NPs modulate AβO neurotoxicity across various experimental systems, we aim to provide valuable insights into novel therapeutic strategies targeting AβOs in AD.
Collapse
Affiliation(s)
| | | | - Yraima Cordeiro
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-900, Brazil
| |
Collapse
|
33
|
Uysal İ, Özden F, Özkeskin M, Benzer Z, Işık Eİ. Exercise Barriers in Older Individuals with Alzheimer's Disease: A Cross-Sectional Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1510. [PMID: 39336551 PMCID: PMC11434187 DOI: 10.3390/medicina60091510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Background and Objectives: Defining the exercise habits of individuals with Alzheimer's Disease (AD) may help to determine optimal rehabilitation programs. This study aimed to investigate the physical and psychological parameters associated with exercise barriers in older individuals with AD, with the goal of informing more effective rehabilitation programs. Materials and Methods: A cross-sectional prospective study was conducted with 50 individuals with AD. The individuals were evaluated with the Exercise Benefit/Barriers Scale (EBBS), the Mini-Mental State Examination (MMSE), the Five Times Sit to Stand Test (FTSTS), the Barthel Index (BI), the Tampa Scale for Kinesiophobia (TSK), and the Hospital Anxiety and Depression Scale (HADS). Results: There was a significant positive correlation between age with EBBS-Exercise Barriers (r = 0.308; p = 0.029) and EBBS-Total Score (r = 0.295; p = 0.038). There were significant negative correlations between the time of diagnosis with EBBS-Exercise Benefits (r = -0.569; p = 0.000), EBBS-Exercise Barriers (r = -0.324; p = 0.022), and EBBS-Total Score (r = -0.508; p = 0.000). There was a positive correlation between MMSE and EBBS-Exercise Benefits (r = 0.465; p = 0.001), EBBS-Exercise Barriers (r = 0.471; p = 0.001) and EBBS-Total Score (r = 0.519; p = 0.000). There were significant positive correlations between FTSTS and EBBS-Exercise Barriers (r = 0.340; p = 0.016), and EBBS-Total Score (r = 0.280; p = 0.049). There were positive correlations between BI and EBBS-Exercise Benefits (r = 0.362; p = 0.010), EBBS-Exercise Barriers (r = 0.377; p = 0.007), and EBBS-Total Score (r = 0.405; p = 0.004). Conclusions: Exercise barriers/benefits were associated with cognition and post-diagnosis duration in individuals with AD. Individuals with lower physical function had lower exercise perception. In addition, living with relatives or caregivers led to better exercise benefit scores.
Collapse
Affiliation(s)
- İsmail Uysal
- Department of Health Care Services, Fethiye Vocational School of Health Services, Muğla Sıtkı Koçman University, 48200 Muğla, Turkey
| | - Fatih Özden
- Department of Health Care Services, Köyceğiz Vocational School of Health Services, Muğla Sıtkı Koçman University, 48800 Muğla, Turkey
| | - Mehmet Özkeskin
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Ege University, 35520 İzmir, Turkey
| | - Zehra Benzer
- Department of Physiotherapy and Rehabilitation, Institute of Health Sciences, Ege University, 35520 İzmir, Turkey
| | - Emir İbrahim Işık
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Çukurova University, 01330 Adana, Turkey
| |
Collapse
|
34
|
Yutani R, Venketaraman V, Sheren N. Treatment of Acute and Long-COVID, Diabetes, Myocardial Infarction, and Alzheimer's Disease: The Potential Role of a Novel Nano-Compound-The Transdermal Glutathione-Cyclodextrin Complex. Antioxidants (Basel) 2024; 13:1106. [PMID: 39334765 PMCID: PMC11429141 DOI: 10.3390/antiox13091106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Oxidative stress (OS) occurs from excessive reactive oxygen species or a deficiency of antioxidants-primarily endogenous glutathione (GSH). There are many illnesses, from acute and post-COVID-19, diabetes, myocardial infarction to Alzheimer's disease, that are associated with OS. These dissimilar illnesses are, in order, viral infections, metabolic disorders, ischemic events, and neurodegenerative disorders. Evidence is presented that in many illnesses, (1) OS is an early initiator and significant promotor of their progressive pathophysiologic processes, (2) early reduction of OS may prevent later serious and irreversible complications, (3) GSH deficiency is associated with OS, (4) GSH can likely reduce OS and restore adaptive physiology, (5) effective administration of GSH can be accomplished with a novel nano-product, the GSH/cyclodextrin (GC) complex. OS is an overlooked pathological process of many illnesses. Significantly, with the GSH/cyclodextrin (GC) complex, therapeutic administration of GSH is now available to reduce OS. Finally, rigorous prospective studies are needed to confirm the efficacy of this therapeutic approach.
Collapse
Affiliation(s)
- Ray Yutani
- Department of Family Medicine, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vishwanath Venketaraman
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Nisar Sheren
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
35
|
Tan LJ, Lei WJ, Liu MM, Cai ZD, Jiang HL, Liu R, Li ZR. Discovery of cinnamamide/ester triazole hybrids as potential treatment for Alzheimer's disease. Bioorg Chem 2024; 150:107584. [PMID: 38964146 DOI: 10.1016/j.bioorg.2024.107584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/30/2024] [Accepted: 06/20/2024] [Indexed: 07/06/2024]
Abstract
Developing multitargeted ligands as promising therapeutics for Alzheimer's disease (AD) has been considered important. Herein, a novel class of cinnamamide/ester-triazole hybrids with multifaceted effects on AD was developed based on the multitarget-directed ligands strategy. Thirty-seven cinnamamide/ester-triazole hybrids were synthesized, with most exhibiting significant inhibitory activity against Aβ-induced toxicity at a single concentration in vitro. The most optimal hybrid compound 4j inhibited copper-induced Aβ toxicity in AD cells. its action was superior to that of donepezil and memantine. It also moderately inhibited intracellular AChE activity and presented favorable bioavailability and blood-brain barrier penetration with low toxicity in vivo. Of note, it ameliorated cognitive impairment, neuronal degeneration, and Aβ deposition in Aβ1-42-injured mice. Mechanistically, the compound regulated APP processing by promoting the ADAM10-associated nonamyloidogenic signaling and inhibiting the BACE1-mediated amyloidogenic pathway. Moreover, it suppressed intracellular AChE activity and tau phosphorylation. Therefore, compound 4j may be a promising multitargeted active molecule against AD.
Collapse
Affiliation(s)
- Lin-Jie Tan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Wen-Ju Lei
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Mi-Min Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Zhong-Di Cai
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hai-Lun Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Rui Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Zhuo-Rong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
36
|
Khare N, Barot M, Singh S, Jain T. Network Pharmacology Reveals Key Targets and Pathways of Madhuca longifolia for Potential Alzheimer's Disease Treatment. Cell Biochem Biophys 2024; 82:2727-2746. [PMID: 39009828 DOI: 10.1007/s12013-024-01389-4] [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] [Accepted: 06/25/2024] [Indexed: 07/17/2024]
Abstract
Madhuca longifolia, commonly known as the mahua tree, has been traditionally used in medicine due to its anti-inflammatory, anti-diabetic, and antimicrobial properties. Its active compounds help in managing diabetes, alleviating cognitive impairment associated with Alzheimer's disease. Nonetheless, the exact neuroprotective mechanism of Madhuca longifolia against Alzheimer's disease remains unclear. This study looked into possible methods by which Madhuca longifolia protects against Alzheimer's disease using network pharmacology, molecular docking and molecular dynamic simulations studies. By applying pre-screening of active constituents, target prediction, Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment analysis, our study found that Madhuca longifolia is related to eight active ingredients (Ascorbic acid, Riboflavin, Pantothenic acid, (4 R)-2beta,3beta,23-trihydroxy-oleana-5,12-dien-28-oic acid, Quercetin, Nicotinic acid, Bassiaic acid Thiamine) and 272 common gene targets, with significant involvement in pathways such as PI3K-Akt signaling and neuroactive ligand-receptor interaction. Network analysis demonstrated how Madhuca longifolia can prevent AD by modifying important signalling networks, which may be one of the molecular mechanisms driving the plant's effectiveness against the disease. Molecular docking studies revealed that there were robust binding abilities of Quercetin, Riboflavin and Pantothenic acid to key target proteins AKT1, JUN, and STAT3. Later, molecular dynamic simulations was done to examine the successful activity of the active compounds against potential targets, and it was found that AKT1 and AKT1-Quercetin complex became stable at 260 ps. It may be seen through the study that quercetin may act as a good inhibitor for treatment. This thorough investigation provides a strong basis for future research and development efforts by advancing our understanding of Madhuca longifolia medicinal potential in Alzheimer's disease.
Collapse
Affiliation(s)
- Noopur Khare
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India
- Bhai Gurdas Institute of Engineering and Technology, Sangrur, Punjab, India
| | - Megha Barot
- Department of Environmental Science, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Sachidanand Singh
- Department of Biotechnology, School of Energy and Technology, Pandit Deendayal Energy University, Gandhinagar, Gujarat, India
| | - Tanvi Jain
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India.
| |
Collapse
|
37
|
Foroughi S, Shahanipour K, Monajemi R, Ahadi AM. Investigating the effects of Thymus vulgaris essential oil, Allium cepa extract, and their active compounds (thymol and quercetin) on expression profile of genes related to Alzheimer's disease in PC12 model cell. Brain Res 2024; 1838:148966. [PMID: 38688382 DOI: 10.1016/j.brainres.2024.148966] [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: 01/04/2024] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
Thymus vulgaris and Allium cepa are plants with great medicinal importance. Thymol monoterpene and quercetin, which are present in these plants, have anti-Alzheimer's and antioxidant effects. The objectives of this research were investigating the effects of these compounds on the pathogenesis and progress of Alzheimer's disease in cells modeled by formaldehyde. MTT, flow cytometry, and RT-PCR were used to investigate the toxicity, survival rate and apoptosis of the cells, and the expression level of PP2A, GSK3, NMDAR, BACE1, and APP genes, respectively. Also, the total antioxidant capacity of the modeled cells was measured. The results showed that the two compounds as well as the plants extract and essential oil were able to increase the percentage of cell survival; among them, Thymus vulgaris essential oil had the greatest effect (93.55316 % in 48 h exposure). In addition, quercetin was able to reduce the rate of apoptosis in Alzheimer's cells (4.73 %) which was greater than the effects of other compounds. In general, the essential oil of Thymus vulgaris compared to thymol; and quercetin compared to Allium cepa extract showed more improving effects on the expression of genes involved in the disease. All four compounds increased the antioxidant capacity of the modeled cells compared to the control group, and these effects were almost equal between the compounds. According to the obtained results, both plants, especially Thymus vulgaris can be proposed as candidates to be included in the diet of Alzheimer's patients. In addition, polyphenols thymol and quercetin as derivates from the studied plants can be used in new drugs development for Alzheimer's disease, with greater safety than currently used drugs. These results are significant because most of the drug for Alzheimer's treatments such as cholinesterases (e.g. rivastigmine and donepezil) and memantine are chemically based and have many side effects.
Collapse
Affiliation(s)
- Saba Foroughi
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Kahin Shahanipour
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Isfahan, Iran.
| | - Ramesh Monajemi
- Department of Biology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Ali Mohammad Ahadi
- Department of Genetics, Faculty of Science, Shahrekord University, ShahreKord, Iran
| |
Collapse
|
38
|
Zhang J, Zhang Y, Wang J, Xia Y, Zhang J, Chen L. Recent advances in Alzheimer's disease: Mechanisms, clinical trials and new drug development strategies. Signal Transduct Target Ther 2024; 9:211. [PMID: 39174535 PMCID: PMC11344989 DOI: 10.1038/s41392-024-01911-3] [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: 11/09/2023] [Revised: 03/18/2024] [Accepted: 07/02/2024] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) stands as the predominant form of dementia, presenting significant and escalating global challenges. Its etiology is intricate and diverse, stemming from a combination of factors such as aging, genetics, and environment. Our current understanding of AD pathologies involves various hypotheses, such as the cholinergic, amyloid, tau protein, inflammatory, oxidative stress, metal ion, glutamate excitotoxicity, microbiota-gut-brain axis, and abnormal autophagy. Nonetheless, unraveling the interplay among these pathological aspects and pinpointing the primary initiators of AD require further elucidation and validation. In the past decades, most clinical drugs have been discontinued due to limited effectiveness or adverse effects. Presently, available drugs primarily offer symptomatic relief and often accompanied by undesirable side effects. However, recent approvals of aducanumab (1) and lecanemab (2) by the Food and Drug Administration (FDA) present the potential in disrease-modifying effects. Nevertheless, the long-term efficacy and safety of these drugs need further validation. Consequently, the quest for safer and more effective AD drugs persists as a formidable and pressing task. This review discusses the current understanding of AD pathogenesis, advances in diagnostic biomarkers, the latest updates of clinical trials, and emerging technologies for AD drug development. We highlight recent progress in the discovery of selective inhibitors, dual-target inhibitors, allosteric modulators, covalent inhibitors, proteolysis-targeting chimeras (PROTACs), and protein-protein interaction (PPI) modulators. Our goal is to provide insights into the prospective development and clinical application of novel AD drugs.
Collapse
Affiliation(s)
- Jifa Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yinglu Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, TN, USA
| | - Yilin Xia
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxian Zhang
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lei Chen
- Department of Neurology, Laboratory of Neuro-system and Multimorbidity and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
39
|
Ramakrishna K, Karuturi P, Siakabinga Q, T A G, Krishnamurthy S, Singh S, Kumari S, Kumar GS, Sobhia ME, Rai SN. Indole-3 Carbinol and Diindolylmethane Mitigated β-Amyloid-Induced Neurotoxicity and Acetylcholinesterase Enzyme Activity: In Silico, In Vitro, and Network Pharmacology Study. Diseases 2024; 12:184. [PMID: 39195183 DOI: 10.3390/diseases12080184] [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: 07/17/2024] [Revised: 08/02/2024] [Accepted: 08/10/2024] [Indexed: 08/29/2024] Open
Abstract
Background: Alzheimer's disease (AD) is a neurodegenerative disease characterized by beta-amyloid (Aβ) deposition and increased acetylcholinesterase (AchE) enzyme activities. Indole 3 carbinol (I3C) and diindolylmethane (DIM) are reported to have neuroprotective activities against various neurological diseases, including ischemic stroke, Parkinson's disease, neonatal asphyxia, depression, stress, neuroinflammation, and excitotoxicity, except for AD. In the present study, we have investigated the anti-AD effects of I3C and DIM. Methods: Docking and molecular dynamic studies against AchE enzyme and network pharmacological studies were conducted for I3C and DIM. I3C and DIM's neuroprotective effects against self and AchE-induced Aβ aggregation were investigated. The neuroprotective effects of I3C and DIM against Aβ-induced neurotoxicity were assessed in SH-S5Y5 cells by observing cell viability and ROS. Results: Docking studies against AchE enzyme with I3C and DIM show binding efficiency of -7.0 and -10.3, respectively, and molecular dynamics studies revealed a better interaction and stability between I3C and AchE and DIM and AchE. Network pharmacological studies indicated that I3C and DIM interacted with several proteins involved in the pathophysiology of AD. Further, I3C and DIM significantly inhibited the AchE (IC50: I3C (18.98 µM) and DIM (11.84 µM)) and self-induced Aβ aggregation. Both compounds enhanced the viability of SH-S5Y5 cells that are exposed to Aβ and reduced ROS. Further, I3C and DIM show equipotential neuroprotection when compared to donepezil. Conclusions: Our findings indicate that both I3C and DIM show anti-AD effects by inhibiting the Aβ induced neurotoxicity and AchE activities.
Collapse
Affiliation(s)
- Kakarla Ramakrishna
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Green Fields, Vaddeswaram, Guntur 522302, Andhra Pradesh, India
| | - Praditha Karuturi
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Green Fields, Vaddeswaram, Guntur 522302, Andhra Pradesh, India
| | - Queen Siakabinga
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Green Fields, Vaddeswaram, Guntur 522302, Andhra Pradesh, India
| | - Gajendra T A
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, IIT BHU, Varanasi 221005, Uttar Pradesh, India
| | - Sairam Krishnamurthy
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, IIT BHU, Varanasi 221005, Uttar Pradesh, India
| | - Shreya Singh
- SBS College of Pharmacy, Malwan, Fatehpur 212664, Uttar Pradesh, India
| | - Sonia Kumari
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Mohali 160062, Punjab, India
| | - G Siva Kumar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Mohali 160062, Punjab, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Mohali 160062, Punjab, India
| | - Sachchida Nand Rai
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
40
|
Rahimi Darehbagh R, Seyedoshohadaei SA, Ramezani R, Rezaei N. Stem cell therapies for neurological disorders: current progress, challenges, and future perspectives. Eur J Med Res 2024; 29:386. [PMID: 39054501 PMCID: PMC11270957 DOI: 10.1186/s40001-024-01987-1] [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: 05/29/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024] Open
Abstract
Stem cell-based therapies have emerged as a promising approach for treating various neurological disorders by harnessing the regenerative potential of stem cells to restore damaged neural tissue and circuitry. This comprehensive review provides an in-depth analysis of the current state of stem cell applications in primary neurological conditions, including Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), stroke, spinal cord injury (SCI), and other related disorders. The review begins with a detailed introduction to stem cell biology, discussing the types, sources, and mechanisms of action of stem cells in neurological therapies. It then critically examines the preclinical evidence from animal models and early human trials investigating the safety, feasibility, and efficacy of different stem cell types, such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs), neural stem cells (NSCs), and induced pluripotent stem cells (iPSCs). While ESCs have been studied extensively in preclinical models, clinical trials have primarily focused on adult stem cells such as MSCs and NSCs, as well as iPSCs and their derivatives. We critically assess the current state of research for each cell type, highlighting their potential applications and limitations in different neurological conditions. The review synthesizes key findings from recent, high-quality studies for each neurological condition, discussing cell manufacturing, delivery methods, and therapeutic outcomes. While the potential of stem cells to replace lost neurons and directly reconstruct neural circuits is highlighted, the review emphasizes the critical role of paracrine and immunomodulatory mechanisms in mediating the therapeutic effects of stem cells in most neurological disorders. The article also explores the challenges and limitations associated with translating stem cell therapies into clinical practice, including issues related to cell sourcing, scalability, safety, and regulatory considerations. Furthermore, it discusses future directions and opportunities for advancing stem cell-based treatments, such as gene editing, biomaterials, personalized iPSC-derived therapies, and novel delivery strategies. The review concludes by emphasizing the transformative potential of stem cell therapies in revolutionizing the treatment of neurological disorders while acknowledging the need for rigorous clinical trials, standardized protocols, and multidisciplinary collaboration to realize their full therapeutic promise.
Collapse
Affiliation(s)
- Ramyar Rahimi Darehbagh
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Nanoclub Elites Association, Tehran, Iran
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Universal Scientific Education and Research Network (USERN), Sanandaj, Kurdistan, Iran
| | | | - Rojin Ramezani
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
Stasiak A, Honkisz-Orzechowska E, Gajda Z, Wagner W, Popiołek-Barczyk K, Kuder KJ, Latacz G, Juszczak M, Woźniak K, Karcz T, Szczepańska K, Jóźwiak-Bębenista M, Kieć-Kononowicz K, Łażewska D. AR71, Histamine H 3 Receptor Ligand-In Vitro and In Vivo Evaluation (Anti-Inflammatory Activity, Metabolic Stability, Toxicity, and Analgesic Action). Int J Mol Sci 2024; 25:8035. [PMID: 39125607 PMCID: PMC11311998 DOI: 10.3390/ijms25158035] [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: 06/21/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
The future of therapy for neurodegenerative diseases (NDs) relies on new strategies targeting multiple pharmacological pathways. Our research led to obtaining the compound AR71 [(E)-3-(3,4,5-trimethoxyphenyl)-1-(4-(3-(piperidin-1-yl)propoxy)phenyl)prop-2-en-1-one], which has high affinity for human H3R (Ki = 24 nM) and selectivity towards histamine H1 and H4 receptors (Ki > 2500 nM), and showed anti-inflammatory activity in a model of lipopolysaccharide-induced inflammation in BV-2 cells. The presented tests confirmed its antagonist/inverse agonist activity profile and good metabolic stability while docking studies showed the binding mode to histamine H1, H3, and H4 receptors. In in vitro tests, cytotoxicity was evaluated at three cell lines (neuroblastoma, astrocytes, and human peripheral blood mononuclear cells), and a neuroprotective effect was observed in rotenone-induced toxicity. In vivo experiments in a mouse neuropathic pain model demonstrated the highest analgesic effects of AR71 at the dose of 20 mg/kg body weight. Additionally, AR71 showed antiproliferative activity in higher concentrations. These findings suggest the need for further evaluation of AR71's therapeutic potential in treating ND and CNS cancer using animal experimental models.
Collapse
Affiliation(s)
- Anna Stasiak
- Department of Hormone Biochemistry, Faculty of Medicine, Medical University of Lodz, Żeligowskiego 7/9 Str., 90-752 Łódź, Poland
| | - Ewelina Honkisz-Orzechowska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College in Kraków, Medyczna 9 Str., 30-688 Kraków, Poland
| | - Zbigniew Gajda
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College in Kraków, Medyczna 9 Str., 30-688 Kraków, Poland
| | - Waldemar Wagner
- Department of Hormone Biochemistry, Faculty of Medicine, Medical University of Lodz, Żeligowskiego 7/9 Str., 90-752 Łódź, Poland
- Laboratory of Cellular Immunology, Institute of Medical Biology of Polish Academy of Sciences, 106 Lodowa Str., 93-232 Łódź, Poland
| | - Katarzyna Popiołek-Barczyk
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Str., 31-343 Kraków, Poland
| | - Kamil J. Kuder
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College in Kraków, Medyczna 9 Str., 30-688 Kraków, Poland
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College in Kraków, Medyczna 9 Str., 30-688 Kraków, Poland
| | - Michał Juszczak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 Str., 90-236 Łódź, Poland
| | - Katarzyna Woźniak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 Str., 90-236 Łódź, Poland
| | - Tadeusz Karcz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College in Kraków, Medyczna 9 Str., 30-688 Kraków, Poland
| | - Katarzyna Szczepańska
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12 Str., 31-343 Kraków, Poland
| | - Marta Jóźwiak-Bębenista
- Department of Pharmacology and Toxicology, Medical University of Lodz, Żeligowskiego 7/9 Str., 90-752 Łódź, Poland
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College in Kraków, Medyczna 9 Str., 30-688 Kraków, Poland
| | - Dorota Łażewska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College in Kraków, Medyczna 9 Str., 30-688 Kraków, Poland
| |
Collapse
|
42
|
Chiricosta L, Minuti A, Gugliandolo A, Salamone S, Pollastro F, Mazzon E, Artimagnella O. Cannabinerol Prevents Endoplasmic Reticulum and Mitochondria Dysfunctions in an In Vitro Model of Alzheimer's Disease: A Network-Based Transcriptomic Analysis. Cells 2024; 13:1012. [PMID: 38920643 PMCID: PMC11201759 DOI: 10.3390/cells13121012] [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: 04/09/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Neurodegenerative disorders are affecting millions of people worldwide, impacting the healthcare system of our society. Among them, Alzheimer's disease (AD) is the most common form of dementia, characterized by severe cognitive impairments. Neuropathological hallmarks of AD are β-amyloid (Aβ) plaques and neurofibrillary tangles, as well as endoplasmic reticulum and mitochondria dysfunctions, which finally lead to apoptosis and neuronal loss. Since, to date, there is no definitive cure, new therapeutic and prevention strategies are of crucial importance. In this scenario, cannabinoids are deeply investigated as promising neuroprotective compounds for AD. In this study, we evaluated the potential neuroprotective role of cannabinerol (CBNR) in an in vitro cellular model of AD via next-generation sequencing. We observed that CBNR pretreatment counteracts the Aβ-induced loss of cell viability of differentiated SH-SY5Y cells. Moreover, a network-based transcriptomic analysis revealed that CBNR restores normal mitochondrial and endoplasmic reticulum functions in the AD model. Specifically, the most important genes regulated by CBNR are related mainly to oxidative phosphorylation (COX6B1, OXA1L, MT-CO2, MT-CO3), protein folding (HSPA5) and degradation (CUL3, FBXW7, UBE2D1), and glucose (G6PC3) and lipid (HSD17B7, ERG28, SCD) metabolism. Therefore, these results suggest that CBNR could be a new neuroprotective agent helpful in the prevention of AD dysfunctions.
Collapse
Affiliation(s)
- Luigi Chiricosta
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Aurelio Minuti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Osvaldo Artimagnella
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
43
|
Yang Y, Qiu L. Research Progress on the Pathogenesis, Diagnosis, and Drug Therapy of Alzheimer's Disease. Brain Sci 2024; 14:590. [PMID: 38928590 PMCID: PMC11201671 DOI: 10.3390/brainsci14060590] [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: 05/15/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
As the population ages worldwide, Alzheimer's disease (AD), the most prevalent kind of neurodegenerative disorder among older people, has become a significant factor affecting quality of life, public health, and economies. However, the exact pathogenesis of Alzheimer's remains elusive, and existing highly recognized pathogenesis includes the amyloid cascade hypothesis, Tau neurofibrillary tangles hypothesis, and neuroinflammation hypothesis. The major diagnoses of Alzheimer's disease include neuroimaging positron emission computed tomography, magnetic resonance imaging, and cerebrospinal fluid molecular diagnosis. The therapy of Alzheimer's disease primarily relies on drugs, and the approved drugs on the market include acetylcholinesterase drugs, glutamate receptor antagonists, and amyloid-β monoclonal antibodies. Still, the existing drugs can only alleviate the symptoms of the disease and cannot completely reverse it. This review aims to summarize existing research results on Alzheimer's disease pathogenesis, diagnosis, and drug therapy, with the objective of facilitating future research in this area.
Collapse
Affiliation(s)
- Yixuan Yang
- College of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Lina Qiu
- College of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
- Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
44
|
Womack CL, Perkins A, Arnold JM. Cognitive Impairment in the Primary Care Clinic. Prim Care 2024; 51:233-251. [PMID: 38692772 DOI: 10.1016/j.pop.2024.02.010] [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: 05/03/2024]
Abstract
Cognitive impairment is a common problem in the geriatric population and is characterized by variable symptoms of memory difficulties, executive dysfunction, language or visuospatial problems, and behavioral changes. It is imperative that primary care clinicians recognize and differentiate the variable symptoms associated with cognitive impairment from changes attributable to normal aging or secondary to other medical conditions. A thorough evaluation for potentially reversible causes of dementia is required before diagnosis with a neurodegenerative dementia. Other abnormal neurologic findings, rapid progression, or early age of onset are red flags that merit referral to neurology for more specialized evaluation and treatment.
Collapse
Affiliation(s)
- Cindy L Womack
- Department of Neurology, Neuroscience Institute, Southern Illinois University School of Medicine, 751 North Rutledge Street, PO 19643, Springfield, IL 62794, USA
| | - Andrea Perkins
- Department of Neurology, Neuroscience Institute, Southern Illinois University School of Medicine, 751 North Rutledge Street, PO 19643, Springfield, IL 62794, USA
| | - Jennifer M Arnold
- Department of Neurology, Neuroscience Institute, Southern Illinois University School of Medicine, 751 North Rutledge Street, PO 19643, Springfield, IL 62794, USA.
| |
Collapse
|
45
|
Gao S, Fan C, Wang Y, Yang W, Jiang H. LncRNA ENST00000440246.1 Promotes Alzheimer's Disease Progression by Targeting PP2A. Biochem Genet 2024; 62:2100-2116. [PMID: 37856039 DOI: 10.1007/s10528-023-10552-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023]
Abstract
Alzheimer's disease (AD) is an extremely prevalent neurodegenerative disease. Long noncoding RNAs (lncRNAs) play pivotal roles in the regulation of AD. However, the function of most lncRNAs in AD remains to be elucidated. In this study, the effects of lncRNA ENST00000440246.1 on the biological characteristics of AD were explored. Differentially expressed lncRNAs in AD were identified through bioinformatics analysis and peripheral blood from thirty AD patients was collected to verify the expression of these lncRNAs by quantitative real-time polymerase chain reaction (RT-qPCR). The correlations between lncRNAs and the Mini-Mental State Examination (MMSE) or the Montreal Cognitive Assessment (MoCA) were assessed by Pearson's correlation analysis. Immunofluorescence (IF), Cell Counting Kit-8 (CCK-8) and flow cytometry assays were conducted to evaluate the biological effect of ENST00000440246.1 and protein phosphatase 2 A (PP2A) in SK-N-SH cells. Gene expression at the protein and mRNA levels was analyzed by Western blotting and RT-qPCR. The interaction between PP2A and ENST00000440246.1 was confirmed by IntaRNA and RNA pulldown assays. ENST00000440246.1 was upregulated and significantly negatively correlated with the MMSE and MoCA scores and the overexpression of ENST00000440246.1 inhibited cell proliferation and facilitated apoptosis and Aβ expression in SK-N-SH cells. Mechanistically, ENST00000440246.1 targeted PP2A and regulated AD-related gene expression. The silencing of ENST00000440246.1 had the opposite effect. Furthermore, PP2A overexpression reversed the influence of ENST00000440246.1 overexpression in SK-N-SH cells. In conclusion, ENST00000440246.1 could promote AD progression by targeting PP2A, which indicates that ENST00000440246.1 has the potential to be a diagnostic target in AD.
Collapse
Affiliation(s)
- Shang Gao
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China
| | - Chang Fan
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China
| | - Yongzhong Wang
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China
- Department of pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China
| | - Wenming Yang
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China.
- Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China.
| | - Hui Jiang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China.
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230031, Anhui, China.
| |
Collapse
|
46
|
Sequeira L, Benfeito S, Fernandes C, Lima I, Peixoto J, Alves C, Machado CS, Gaspar A, Borges F, Chavarria D. Drug Development for Alzheimer's and Parkinson's Disease: Where Do We Go Now? Pharmaceutics 2024; 16:708. [PMID: 38931832 PMCID: PMC11206728 DOI: 10.3390/pharmaceutics16060708] [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: 04/04/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a set of progressive, chronic, and incurable diseases characterized by the gradual loss of neurons, culminating in the decline of cognitive and/or motor functions. Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common NDs and represent an enormous burden both in terms of human suffering and economic cost. The available therapies for AD and PD only provide symptomatic and palliative relief for a limited period and are unable to modify the diseases' progression. Over the last decades, research efforts have been focused on developing new pharmacological treatments for these NDs. However, to date, no breakthrough treatment has been discovered. Hence, the development of disease-modifying drugs able to halt or reverse the progression of NDs remains an unmet clinical need. This review summarizes the major hallmarks of AD and PD and the drugs available for pharmacological treatment. It also sheds light on potential directions that can be pursued to develop new, disease-modifying drugs to treat AD and PD, describing as representative examples some advances in the development of drug candidates targeting oxidative stress and adenosine A2A receptors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Fernanda Borges
- CIQUP-IMS—Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS—Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| |
Collapse
|
47
|
Morán-Serradilla C, Plano D, Sanmartín C, Sharma AK. Selenization of Small Molecule Drugs: A New Player on the Board. J Med Chem 2024; 67:7759-7787. [PMID: 38716896 DOI: 10.1021/acs.jmedchem.3c02426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
There is an urgent need to develop safer and more effective modalities for the treatment of a wide range of pathologies due to the increasing rates of drug resistance, undesired side effects, poor clinical outcomes, etc. Throughout the years, selenium (Se) has attracted a great deal of attention due to its important role in human health. Besides, a growing body of work has unveiled that the inclusion of Se motifs into a great number of molecules is a promising strategy for obtaining novel therapeutic agents. In the current Perspective, we have gathered the most recent literature related to the incorporation of different Se moieties into the scaffolds of a wide range of known drugs and their feasible pharmaceutical applications. In addition, we highlight different representative examples as well as provide our perspective on Se drugs and the possible future directions, promises, opportunities, and challenges of this ground-breaking area of research.
Collapse
Affiliation(s)
| | - Daniel Plano
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, Pamplona E-31008, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, Pamplona E-31008, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, United States
- Penn State Cancer Institute, 400 University Drive,Hershey, Pennsylvania 17033, United States
| |
Collapse
|
48
|
Zelek WM, Bevan RJ, Morgan BP. Targeting terminal pathway reduces brain complement activation, amyloid load and synapse loss, and improves cognition in a mouse model of dementia. Brain Behav Immun 2024; 118:355-363. [PMID: 38485063 DOI: 10.1016/j.bbi.2024.03.017] [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: 12/05/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024] Open
Abstract
Complement is dysregulated in the brain in Alzheimer's Disease and in mouse models of Alzheimer's disease. Each of the complement derived effectors, opsonins, anaphylatoxins and membrane attack complex (MAC), have been implicated as drivers of disease but their relative contributions remain unclarified. Here we have focussed on the MAC, a lytic and pro-inflammatory effector, in the AppNL-G-F mouse amyloidopathy model. To test the role of MAC, we back-crossed to generate AppNL-G-F mice deficient in C7, an essential MAC component. C7 deficiency ablated MAC formation, reduced synapse loss and amyloid load and improved cognition compared to complement-sufficient AppNL-G-F mice at 8-10 months age. Adding back C7 caused increased MAC formation in brain and an acute loss of synapses in C7-deficient AppNL-G-F mice. To explore whether C7 was a viable therapeutic target, a C7-blocking monoclonal antibody was administered systemically for one month in AppNL-G-F mice aged 8-9 months. Treatment reduced brain MAC and amyloid deposition, increased synapse density and improved cognitive performance compared to isotype control-treated AppNL-G-F mice. The findings implicate MAC as a driver of pathology and highlight the potential for complement inhibition at the level of MAC as a therapy in Alzheimer's disease.
Collapse
Affiliation(s)
- Wioleta M Zelek
- UK Dementia Research Institute Cardiff and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, Wales CF14 4XN, United Kingdom.
| | - Ryan J Bevan
- UK Dementia Research Institute Cardiff and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, Wales CF14 4XN, United Kingdom
| | - Bryan Paul Morgan
- UK Dementia Research Institute Cardiff and Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, Wales CF14 4XN, United Kingdom.
| |
Collapse
|
49
|
Belozor OS, Vasilev A, Mileiko AG, Mosina LD, Mikhailov IG, Ox DA, Boitsova EB, Shuvaev AN, Teschemacher AG, Kasparov S, Shuvaev AN. Memantine suppresses the excitotoxicity but fails to rescue the ataxic phenotype in SCA1 model mice. Biomed Pharmacother 2024; 174:116526. [PMID: 38574621 DOI: 10.1016/j.biopha.2024.116526] [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: 12/27/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a debilitating neurodegenerative disorder of the cerebellum and brainstem. Memantine has been proposed as a potential treatment for SCA1. It blocks N-methyl-D-aspartate (NMDA) receptors on neurons, reduces excitotoxicity and decreases neurodegeneration in Alzheimer models. However, in cerebellar neurodegenerative diseases, the potential value of memantine is still unclear. We investigated the effects of memantine on motor performance and synaptic transmission in the cerebellum in a mouse model where mutant ataxin 1 is specifically targeted to glia. Lentiviral vectors (LVV) were used to express mutant ataxin 1 selectively in Bergmann glia (BG). In mice transduced with the mutant ataxin 1, chronic treatment with memantine improved motor activity during initial tests, presumably due to preserved BG and Purkinje cell (PC) morphology and numbers. However, mice were unable to improve their rota rod scores during next days of training. Memantine also compromised improvement in the rota rod scores in control mice upon repetitive training. These effects may be due to the effects of memantine on plasticity (LTD suppression) and NMDA receptor modulation. Some effects of chronically administered memantine persisted even after its wash-out from brain slices. Chronic memantine reduced morphological signs of neurodegeneration in the cerebellum of SCA1 model mice. This resulted in an apparent initial reduction of ataxic phenotype, but memantine also affected cerebellar plasticity and ultimately compromised motor learning. We speculate that that clinical application of memantine in SCA1 might be hampered by its ability to suppress NMDA-dependent plasticity in cerebellar cortex.
Collapse
Affiliation(s)
- Olga S Belozor
- Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Partizan Zheleznyak st. 1, Krasnoyarsk 660022, Russia
| | - Alex Vasilev
- JSC «BIOCAD», Svyazi str. 34-A, Strelna, Saint-Petersburg 198515, Russia
| | | | - Lyudmila D Mosina
- Siberian Federal University, Svobodny pr., 79, Krasnoyarsk 660041, Russia
| | - Ilya G Mikhailov
- Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Partizan Zheleznyak st. 1, Krasnoyarsk 660022, Russia; Siberian Federal University, Svobodny pr., 79, Krasnoyarsk 660041, Russia
| | - Darius A Ox
- Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Partizan Zheleznyak st. 1, Krasnoyarsk 660022, Russia; Siberian Federal University, Svobodny pr., 79, Krasnoyarsk 660041, Russia
| | - Elizaveta B Boitsova
- Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Partizan Zheleznyak st. 1, Krasnoyarsk 660022, Russia
| | - Andrey N Shuvaev
- Siberian Federal University, Svobodny pr., 79, Krasnoyarsk 660041, Russia
| | - Anja G Teschemacher
- Department of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Sergey Kasparov
- Department of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Anton N Shuvaev
- Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Partizan Zheleznyak st. 1, Krasnoyarsk 660022, Russia; Siberian Federal University, Svobodny pr., 79, Krasnoyarsk 660041, Russia.
| |
Collapse
|
50
|
Patil N, Dhariwal R, Mohammed A, Wei LS, Jain M. Network pharmacology-based approach to elucidate the pharmacologic mechanisms of natural compounds from Dictyostelium discoideum for Alzheimer's disease treatment. Heliyon 2024; 10:e28852. [PMID: 38644825 PMCID: PMC11033062 DOI: 10.1016/j.heliyon.2024.e28852] [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: 10/27/2023] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/23/2024] Open
Abstract
Alzheimer's disease (AD) is increasingly becoming a major public health concern in our society. While many studies have explored the use of natural polyketides, alkaloids, and other chemical components in AD treatment, there is an urgent need to clarify the concept of multi-target treatment for AD. This study focuses on using network pharmacology approach to elucidate how secondary metabolites from Dictyostelium discoideum affect AD through multi-target or indirect mechanisms. The secondary metabolites produced by D. discoideum during their development were obtained from literature sources and PubChem. Disease targets were selected using GeneCards, DisGeNET, and CTD databases, while compound-based targets were identified through Swiss target prediction and Venn diagrams were used to find intersections between these targets. A network depicting the interplay among disease, drugs, active ingredients, and key target proteins (PPI network) was formed utilizing the STRING (Protein-Protein Interaction Networks Functional Enrichment Analysis) database. To anticipate the function and mechanism of the screened compounds, GO and KEGG enrichment analyses were conducted and visually presented using graphs and bubble charts. After the screening phase, the top interacting targets in the PPI network and the compound with the most active target were chosen for subsequent molecular docking and molecular dynamic simulation studies. This study identified nearly 50 potential targeting genes for each of the screened compounds and revealed multiple signaling pathways. Among these pathways, the inflammatory pathway stood out. COX-2, a receptor associated with neuroinflammation, showed differential expression in various stages of AD, particularly in pyramidal neurons during the early stages of the disease. This increase in COX-2 expression is likely induce by higher levels of IL-1, which is associated with neuritic plaques and microglial cells in AD. Molecular docking investigations demonstrated a strong binding interaction between the terpene compound PQA-11 and the neuroinflammatory receptor COX2, with a substantial binding affinity of -8.4 kcal/mol. Subsequently, a thorough analysis of the docked complex (COX2-PQA11) through Molecular Dynamics Simulation showed lower RMSD, minimal RMSF fluctuations, and a reduced total energy of -291.35 kJ/mol compared to the standard drug. These findings suggest that the therapeutic effect of PQA-11 operates through the inflammatory pathway, laying the groundwork for further in-depth research into the role of secondary metabolites in AD treatment.
Collapse
Affiliation(s)
- Nil Patil
- Cell & Developmental Biology Lab, Research & Development Cell, Parul University, Vadodara, Gujarat, 391760, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, 391760, India
| | - Rupal Dhariwal
- Cell & Developmental Biology Lab, Research & Development Cell, Parul University, Vadodara, Gujarat, 391760, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, 391760, India
| | - Arifullah Mohammed
- Department of Agriculture, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli, 17600, Malaysia
| | - Lee Seong Wei
- Department of Agriculture, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli, 17600, Malaysia
| | - Mukul Jain
- Cell & Developmental Biology Lab, Research & Development Cell, Parul University, Vadodara, Gujarat, 391760, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, 391760, India
| |
Collapse
|