1
|
Liu X, Xu J, Yu C, Dai C, Chen J, Zhong J, Yang Y, Lin H, Chen X, Zhang Q, Dai L, Zhang J, Zha D, Ye ZC. A novel carbamate-based hybrid derivative with anti-neuroinflammatory properties as a selective butyrylcholinesterase inhibitor for Alzheimer's disease therapy. Bioorg Chem 2025; 161:108551. [PMID: 40347768 DOI: 10.1016/j.bioorg.2025.108551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/22/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
Cholinesterase inhibitors (ChEIs) are widely utilized for the symptomatic management of Alzheimer's disease (AD) by enhancing acetylcholine levels to improve cognitive function. Concurrently, neuroinflammation has emerged as a critical factor in AD progression, necessitating therapies that address this pathology. In this study, we designed and synthesized a novel bifunctional cholinesterase inhibitor, (E)-4-(2-(3-(benzyloxy)-4-oxo-4H-pyran-2-yl) vinyl)-1,2-phenylene bis(ethyl(methyl)carbamate) (D40), which combines potent cholinesterase inhibition with robust anti-neuroinflammatory activity. D40 demonstrated potent inhibition of human butyrylcholinesterase (hBuChE), with an IC₅₀ value of 0.59 ± 0.03 μM, significantly outperforming Rivastigmine (IC₅₀ = 3.70 ± 0.96 μM). Molecular docking and molecular dynamics simulations confirmed a stable and selective binding of D40 to the BuChE active site, underpinning its inhibitory profile. Additionally, D40 exhibited strong anti-inflammatory effects, with an IC₅₀ value of 4.55 ± 0.78 μM for suppressing nitric oxide production and demonstrated excellent blood-brain barrier permeability. In vivo studies in aged 5 × FAD mice revealed that D40 significantly reduced neuroinflammation by suppressing pro-inflammatory cytokines and glial activation. Furthermore, D40 mitigated Aβ deposition, promoted neuronal survival, and improved cognitive deficits, while demonstrating a favorable safety profile in acute toxicity evaluations. These findings highlight D40 as a dual-function ChEI capable of providing symptomatic relief and modulating neuroinflammatory pathways associated with AD. With its enhanced cholinesterase inhibition and anti-inflammatory properties, D40 emerges as a promising candidate for the treatment of advanced stages of AD. Acetylcholine deficiency and neuroinflammation as drivers of Alzheimer's disease dually intervened by Compound D40.
Collapse
Affiliation(s)
- Xueyan Liu
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China; Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jiexin Xu
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Chuanyu Yu
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Chaoxian Dai
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jiajing Chen
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jian Zhong
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Yaoyao Yang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Huiling Lin
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Xili Chen
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Qianling Zhang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Luyao Dai
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China
| | - Jing Zhang
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, Fujian Province, China.
| | - Daijun Zha
- School of Pharmacy, Fujian Medical University, Fuzhou 350112, Fujian Province, China.
| | - Zu-Cheng Ye
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350112, Fujian Province, China.
| |
Collapse
|
2
|
Lu Z, Jiang Z, Huang X, Chen Y, Feng L, Mai J, Lao L, Li L, Chen WH, Hu J. Anti-Alzheimer effects of an HDAC6 inhibitor, WY118, alone and in combination of lithium chloride: Synergistic suppression of ferroptosis via the modulation of tau phosphorylation and MAPK signaling. Eur J Pharmacol 2025; 997:177605. [PMID: 40204225 DOI: 10.1016/j.ejphar.2025.177605] [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: 02/06/2025] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder, and current therapies mainly offer symptomatic relief. Given that the pathophysiology of AD is multifaceted, a multimodal therapeutic strategy targeting multiple molecular pathways implicated in AD-related pathogenesis represents a pragmatic avenue for impeding the advancement of AD. In this study, we evaluated the anti-Alzheimer effects of an HDAC6 inhibitor WY118, both alone and in combination with lithium chloride (LiCl), a GSK-3β inhibitor, to synergistically suppress ferroptosis. The combination of compound WY118 and LiCl demonstrated significant synergistic effects in both cellular models of AD induced by glutamate and streptozotocin. The findings suggest that compound WY118, in particular in combination with LiCl, exhibits potent anti-Alzheimer effects by synergistically suppressing ferroptosis. Studies on the mechanism of action indicated that the combination treatment significantly reduced tau phosphorylation and inhibited p38 MAPK signaling. This combination therapy holds promise for developing more effective treatments for AD.
Collapse
Affiliation(s)
- Zhonghui Lu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Zixing Jiang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Xiaoling Huang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Yu Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Luanqi Feng
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Jielin Mai
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Linghui Lao
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Lanqing Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Wen-Hua Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China.
| | - Jinhui Hu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China.
| |
Collapse
|
3
|
Wang SM, Ji WS, Chen LJ, Shan LH, Li X, Gao F, Xu JB. Design, synthesis, and bio-evaluation of C1-aryl galantamine derivatives. Fitoterapia 2025; 183:106535. [PMID: 40204048 DOI: 10.1016/j.fitote.2025.106535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/24/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
For exploring the naturally-inspired anti-AD multifunctional molecules, a series of C1-aryl galantamine derivatives 4a-x were designed and synthesized. The cholinesterase inhibition activity and neuroprotection ability of 4a-x were evaluated. Among them, meta-tbutylbenzene derivative 4b (IC50 = 0.12 ± 0.03 μM) and para-hydroxybenzene derivative 4h (IC50 = 3.86 ± 0.82 μM) exhibited selective inhibition effect against EeAChE and EqBChE, respectively. Compound 4k containing meta-methylthio benzene group exhibited dual inhibitory activity against EeAChE (IC50 = 0.47 ± 0.13 μM) and EqBChE (IC50 = 2.98 ± 0.80 μM). Furthermore, the neuroprotection experiment revealed that eight synthesized derivatives had significant effect to protect SH-SY5Y cells from H2O2-induced damage at a concentration of 25 μM. Notably, the dual AChE and BChE inhibitor 4k also exhibited the good neuroprotective activity, which deserves to be further explored for developing potential multifunctional AD drug.
Collapse
Affiliation(s)
- Su-Mei Wang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| | - Wan-Sheng Ji
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Long-Jiang Chen
- Kelun Pharmaceutical Research Institute, Chengdu 611138, China
| | - Lian-Hai Shan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiaohuan Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Feng Gao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Yibin Institute of Southwest Jiaotong University, Yibin 644000, Sichuan, China.
| | - Jin-Bu Xu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Yibin Institute of Southwest Jiaotong University, Yibin 644000, Sichuan, China.
| |
Collapse
|
4
|
Joshi N, Alavala RR. Identification of novel ligands against GSK-3β: Molecular docking, ADMET filtering, MD simulations, MM-GBSA studies, and DFT analysis. Comput Biol Chem 2025; 118:108478. [PMID: 40300214 DOI: 10.1016/j.compbiolchem.2025.108478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 05/01/2025]
Abstract
Glycogen synthase kinase-3β (GSK-3β) is a key enzyme involved in tau hyperphosphorylation, a key pathological mechanism leading to tau aggregation and thus, Alzheimer's disease. It also has a significant role in neuroinflammation, induction of BACE1, and amyloid-β aggregation. Such crucial pathological events are involved intricately with the Alzheimer's disease pathophysiology which makes this enzyme an attractive drug target. Designing a competitive ATP-site inhibitor of GSK-3β is the approach adopted here in an attempt to target tau pathology. Tetrazole is selected as a scaffold taking into account the previously reported inhibitors bearing nitrogen heterocycles which are at the heart of drug design strategies. A library of disubstituted tetrazole analogs was designed and molecular docking was performed. The top 30 molecules were then passed through several ADMET filters with a special preference given to BBB permeability. The hit molecules obtained after this were analyzed for their amino acid interactions. The docked complexes were used to perform MD simulations for 100 ns and later MM-GBSA studies were performed with staurosporine and co-crystallized ligand as reference molecules. Furthermore, various chemical reactivity parameters and the HOMO-LUMO energy gap were analyzed with DFT studies using B3LYP functional. The best molecule obtained was TD30, revealing a docking score of -167.036, an average RMSD of 2.15 Å for 100 ns simulation time, average ΔGBinding energy of -86.55 ± 4.45 kcal/mol, and an HOMO-LUMO energy gap of 4.044 eV; all corroborating the potential of TD30 for future research and enzyme inhibition assays.
Collapse
Affiliation(s)
- Nachiket Joshi
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - RajaSekhar Reddy Alavala
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India.
| |
Collapse
|
5
|
Ben Zaken K, Bouhnik R, Omer N, Bloch N, Samson AO. Polyoxometalates bind multiple targets involved in Alzheimer's disease. J Biol Inorg Chem 2025; 30:299-309. [PMID: 40119889 PMCID: PMC11965166 DOI: 10.1007/s00775-025-02111-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by brain aggregates of amyloid-β (Aβ) plaques and Tau tangles. Despite extensive research, effective therapy for AD remains elusive. Polyoxometalates (POMs), a class of inorganic compounds with diverse chemical structures and properties, are emerging as potential candidates for AD treatment due to their ability to target key molecular players implicated in disease pathogenesis, such as Aβ, acetylcholinesterase (AChE) and butyryl acetylcholinesterase (BChE). Here, we use molecular docking to predict the binding pose and affinities of POMs to 10 top targets associated with AD. First, we validate our method by replicating experimentally known binding of POMs to Aβ (ΔG = - 9.67 kcal/mol), AChE (ΔG = - 9.39 kcal/mol) and BChE (ΔG = - 10.86 kcal/mol). Then, using this method, we show that POM can also bind β-secretase 1 (BACE1, ΔG = - 10.14 kcal/mol), presenilin 1 (PSEN1, ΔG = - 10.65 kcal/mol), presenilin 2 (PSEN2, ΔG = - 7.94 kcal/mol), Amyloid Precursor Protein (APP, ΔG = - 7.26 kcal/mol), Apolipoprotein E (APOE4, ΔG = - 10.05 kcal/mol), Microtubule-Associated Protein Tau (MAPT, ΔG = - 5.28 kcal/mol) depending on phosphorylation, and α-synuclein (SNCA, ΔG = - 7.64 kcal/mol). Through such binding, POMs offer the potential to mitigate APP cleavage, Aβ oligomer neurotoxicity, Aβ aggregation, thereby attenuating disease progression. Overall, our molecular docking study represents a powerful tool in the discovery of POM-based therapeutics for AD, facilitating the development of novel treatments for AD.
Collapse
Affiliation(s)
- Karin Ben Zaken
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Rivka Bouhnik
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Naama Omer
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Naamah Bloch
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Abraham O Samson
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel.
| |
Collapse
|
6
|
Uliassi E, Bolognesi ML, Milelli A. Targeting Tau Protein with Proximity Inducing Modulators: A New Frontier to Combat Tauopathies. ACS Pharmacol Transl Sci 2025; 8:654-672. [PMID: 40109749 PMCID: PMC11915046 DOI: 10.1021/acsptsci.4c00733] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 03/22/2025]
Abstract
Dysregulation of correct protein tau homeostasis represents the seed for the development of several devastating central nervous system disorders, known as tauopathies, that affect millions of people worldwide. Despite massive public and private support to research funding, these diseases still represent unmet medical needs. In fact, the tau-targeting tools developed to date have failed to translate into the clinic. Recently, taking advantage of the modes that nature uses to mediate the flow of information in cells, researchers have developed a new class of molecules, called proximity-inducing modulators, which exploit spatial proximity to modulate protein function(s) and redirect cellular processes. In this perspective, after a brief discussion about tau protein and the classic tau-targeting approaches, we will discuss the different classes of proximity-inducing modulators developed so far and highlight the applications to modulate tau protein's function and tau-induced toxicity.
Collapse
Affiliation(s)
- Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
| | - Andrea Milelli
- Department for Life Quality Studies, Alma Mater Studiorum - University of Bologna, Corso d'Augusto 237, Rimini 47921, Italy
| |
Collapse
|
7
|
Wang Y, Huang L, Cen X, Liang Y, Chen K. Canonical MAPK signaling in auditory neuropathy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167619. [PMID: 39662753 DOI: 10.1016/j.bbadis.2024.167619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/27/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Auditory neuropathy (AN) is an under-recognized form of hearing loss characterized by lesions in inner hair cells (IHCs), ribbon synapses and spiral ganglion neurons (SGNs). The lack of a targeted therapy for AN has increased the need for a better understanding of the pathogenic mechanism of AN. As mitogen-activated protein kinase (MAPK) signaling is ubiquitous in many biological processes, its alteration may facilitate the pathogenesis of multiple sites in AN. Here, we summaries the characteristics of AN under different molecular bases and first explore the mechanism of MAPK at different lesion sites. Alterations of extracellular signal-regulated kinase (ERK)/MAPK occur in IHCs and SGNs, whereas modulations of p38 and c-Jun NH2-terminal kinase (JNK) were found in ribbon synapses and SGNs. In conclusion, inductive MAPK alterations in the pathogenesis and development of AN are likely to represent a potential therapeutic target to guide the development of treatments.
Collapse
Affiliation(s)
- Yueying Wang
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China
| | - Lusha Huang
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoqing Cen
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China
| | - Yue Liang
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China
| | - Kaitian Chen
- Otorhinolaryngology Hospital, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Institute of Otorhinolaryngology, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
8
|
Taufeeq M, Choudhury A, Hussain A, Alajmi MF, Mohammad T, Shamsi A, Hassan MI. Discovering potential ERK1 inhibitors from natural products for therapeutic targeting of Alzheimer's disease. J Alzheimers Dis 2025; 103:1226-1244. [PMID: 39814427 DOI: 10.1177/13872877241309592] [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: 01/18/2025]
Abstract
BACKGROUND Extracellular signal-regulated kinase 1 (ERK1) belongs to mitogen-activated protein kinases, which are essential for memory formation, cognitive function, and synaptic plasticity. During Alzheimer's disease (AD), ERK1 phosphorylates tau at 15 phosphorylation sites, leading to the formation of neurofibrillary tangles. The overactivation of ERK1 in microglia promotes the release of pro-inflammatory cytokines, which results in neuroinflammation. Additionally, elevated oxidative stress during AD stimulates the ERK1 pathway, leading to neuronal loss. OBJECTIVE Because ERK1 signaling plays a significant role in tau phosphorylation, targeting ERK1 may be therapeutically beneficial by either preventing excessive activation of the signaling pathway or altering its pathway to enhance neuroprotective effects during AD. METHODS This study employed structure-based virtual screening of phytoconstituents from the IMPPAT library. Subsequently, in-depth docking and molecular dynamics (MD) simulation studies were implemented to identify potential ERK1 inhibitors with desirable pharmacological properties. RESULTS Silandrin and Hydroxytuberosone were found to be potential ERK1 inhibitors with higher affinity and specificity than the control molecule Tizaterkib. These compounds specifically bind to the ERK1 substrate binding pocket and interact with crucial residues. Finally, the elucidated compounds with ERK1 were evaluated using an all-atom molecular MD simulation to analyze structural dynamics, structural compactness, hydrogen bond dynamics, principal component analysis, and free energy landscape. CONCLUSIONS The study suggested that Silandrin and Hydroxytuberosone can further be exploited as potential lead molecules for therapeutic development against ERK1-mediated AD.
Collapse
Affiliation(s)
- Mohammad Taufeeq
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
9
|
He D, Zhang X, Su J, Zhang Q, Zhao L, Wu T, Ren H, Jia R, Lei X, Hou W, Sun W, Fan Y, Wang Z. Identification of AS1842856 as a novel small-molecule GSK3α/β inhibitor against Tauopathy by accelerating GSK3α/β exocytosis. Aging Cell 2025; 24:e14336. [PMID: 39287420 PMCID: PMC11709109 DOI: 10.1111/acel.14336] [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: 04/14/2024] [Revised: 07/11/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Glycogen synthase kinase-3α/β (GSK3α/β) is a critical kinase for Tau hyperphosphorylation which contributes to neurodegeneration. Despite the termination of clinical trials for GSK3α/β inhibitors in Alzheimer's disease (AD) treatment, there is a pressing need for novel therapeutic strategies targeting GSK3α/β. Here, we identified the compound AS1842856 (AS), a specific forkhead box protein O1 (FOXO1) inhibitor, reduced intracellular GSK3α/β content in a FOXO1-independent manner. Specifically, AS directly bound to GSK3α/β, promoting its translocation to the multivesicular bodies (MVBs) and accelerating exocytosis, ultimately decreasing intracellular GSK3α/β content. Expectedly, AS treatment effectively suppressed Tau hyperphosphorylation in cells exposed to okadaic acid or expressing the TauP301S mutant. Furthermore, AS was visualized to penetrate the blood-brain barrier (BBB) using an imaging mass microscope. Long-term treatment of AS enhanced cognitive function in P301S transgenic mice by mitigating Tau hyperphosphorylation through downregulation of GSK3α/β expression in the brain. Altogether, AS represents a novel small-molecule GSK3α/β inhibitor that facilitates GSK3α/β exocytosis, holding promise as a therapeutic agent for GSK3α/β hyperactivation-associated disorders.
Collapse
Affiliation(s)
- Da‐Long He
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| | - Xiao‐Yu Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical PhysicsChinese Academy of SciencesDalianChina
| | - Jing‐Yang Su
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| | - Qi Zhang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| | - Ling‐Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| | - Ting‐Yao Wu
- First Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | - Hang Ren
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| | - Rong‐Jun Jia
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| | - Xian‐Fang Lei
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| | - Wen‐Jia Hou
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| | - Wen‐Ge Sun
- Department of RadiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Yong‐Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| | - Zhan‐You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning ProvinceHealth Sciences Institute of China Medical UniversityShenyangChina
| |
Collapse
|
10
|
Goetzl EJ. Current Developments in Alzheimer's Disease. Am J Med 2025; 138:15-20. [PMID: 39197720 DOI: 10.1016/j.amjmed.2024.08.019] [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: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
An increased understanding of the predisposing genetics and complex pathogenic mechanisms of Alzheimer's disease have facilitated delineation of the long preclinical course and re-invigorated the search for disease-modifying treatments. Establishment of accurate blood-based biomarkers has enabled preclinical identification of early disease and permits trials of preventative treatment and quantitative monitoring of therapeutic effects. The broad range of therapeutic possibilities encompasses gene editing, enzyme activators and inhibitors, antisense oligonucleotides, and antagonists of receptors for inflammatory mediators.
Collapse
Affiliation(s)
- Edward J Goetzl
- Department of Medicine, University of California Medical Center, San Francisco.
| |
Collapse
|
11
|
Chen TY, Chen YR, Hsu ML, Liao YT, Wu CH, Yao CA, Yang WC, Lin W, Lin Y. Homoplantaginin Antagonizes N-Methyl-d-aspartate Receptor and Extracellular Signal-Regulated Kinase Signaling in Aβ Oligomers-Induced Neuropathology/Toxicity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:28294-28304. [PMID: 39661093 DOI: 10.1021/acs.jafc.4c07659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Extracts from plants/herbals are great resources of drugs and nutrients. Baicalein, a component present in Scutellaria baicalensis, was previously found to alleviate the abnormal depolarization brought about by Aβ oligomers. We extended this promising outcome by screening baicalein derivatives, and a natural compound named homoplantaginin was pinpointed. In this study, we aimed to investigate the effects of homoplantaginin on animal behavior and explore its neuronal functioning/mechanism. In behavior tests, impairments of novel object recognition and of spatial learning/memory were reversed by homoplantaginin in a J20 Alzheimer's disease (AD) mouse model. Utilizing primary glutamatergic neurons, homoplantaginin was found to prevent the Aβ oligomer-induced increase in ERK phosphorylation. Furthermore, homoplantaginin inhibits both AMPA-insult and NMDA-insult depolarization; this was assessed using DiBAC4(3), a membrane potential sensitive dye. Finally, homoplantaginin blocks both Aβ oligomer-induced and NMDA-induced calcium influx, which was assessed by intracellular calcium measurement using Fura2/AM. Interestingly, homoplantaginin immediately blunts the steady state calcium influx caused by NMDA. Taken together, homoplantaginin is capable of inhibiting Aβ oligomer-induced pathophysiology, in particular at the receptor level. This pure compound has great potential to be developed as a clinical therapeutic drug.
Collapse
Affiliation(s)
- Ting-Yu Chen
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Yi-Ru Chen
- Department of Chemistry, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Ming-Lung Hsu
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Yueh-Ting Liao
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Chia-Hsuan Wu
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Chien-An Yao
- Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Wen-Chin Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei 116325, Taiwan
| | - Yenshou Lin
- Department of Life Science, National Taiwan Normal University, Taipei 116325, Taiwan
| |
Collapse
|
12
|
Tu HJ, Chao MW, Lee CC, Peng CS, Wu YW, Lin TE, Chang YW, Yen SC, Hsu KC, Pan SL, HuangFu WC. Discovering a novel dual specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) inhibitor and its impact on tau phosphorylation and amyloid-β formation. J Enzyme Inhib Med Chem 2024; 39:2418470. [PMID: 39494990 PMCID: PMC11536634 DOI: 10.1080/14756366.2024.2418470] [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: 07/23/2024] [Revised: 09/25/2024] [Accepted: 10/12/2024] [Indexed: 11/05/2024] Open
Abstract
Dual-specificity tyrosine-regulated kinase 1 A (DYRK1A) is crucial in neurogenesis, synaptogenesis, and neuronal functions. Its dysregulation is linked to neurodegenerative disorders like Down syndrome and Alzheimer's disease. Although the development of DYRK1A inhibitors has significantly advanced in recent years, the selectivity of these drugs remains a critical challenge, potentially impeding further progress. In this study, we utilised structure-based virtual screening (SBVS) from NCI library to discover novel DYRK1A inhibitors. The top-ranked compounds were then validated through enzymatic assays to assess their efficacy towards DYRK1A. Among them, NSC361563 emerged as a potent and selective DYRK1A inhibitor. It was shown to decrease tau phosphorylation at multiple sites, thereby enhancing tubulin stability. Moreover, NSC361563 diminished the formation of amyloid β and offered neuroprotective benefits against amyloid β-induced toxicity. Our research highlights the critical role of selective DYRK1A inhibitors in treating neurodegenerative diseases and presents a promising starting point for the development of targeted therapies.
Collapse
Affiliation(s)
- Huang-Ju Tu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Min-Wu Chao
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Institute of Biopharmaceutical Sciences, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
- The Doctoral Program of Clinical and Experimental Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Cheng-Chung Lee
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chao-Shiang Peng
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yi-Wen Wu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wei Chang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung Medical Center, Keelung, Taiwan
| | - Shih-Chung Yen
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong, People’s Republic of China
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shiow-Lin Pan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chun HuangFu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
13
|
Yoon G, Kam MK, Koh YH, Jo C. Palmitoyl-L-carnitine induces tau phosphorylation and mitochondrial dysfunction in neuronal cells. PLoS One 2024; 19:e0313507. [PMID: 39536002 PMCID: PMC11560007 DOI: 10.1371/journal.pone.0313507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline and memory loss, involving mechanisms such as tau hyperphosphorylation and mitochondrial dysfunction. Increasing evidence suggests that age-related alterations in metabolite levels are crucial for the pathogenesis of AD. Here, we analyzed serum metabolites from mice of various ages (2, 4, 14, and 21 months old) using mass spectrometry. We identified palmitoyl-L-carnitine as a key metabolite with significantly increased levels in aged mice. In vitro experiments with SH-SY5Y neuronal cells demonstrated that palmitoyl-L-carnitine treatment enhanced tau phosphorylation, increased mitochondrial fission, and elevated intracellular calcium levels. Furthermore, the increased levels of tau phosphorylation were significantly reduced by the inhibition of GSK-3β, CDK5, and calpain, indicating that tau kinases activated by calcium overload are directly involved in the increase of tau phosphorylation. Considering that mitochondrial fission is related to mitochondrial dysfunction, we propose that the elevated level of serum palmitoyl-L-carnitine during aging contributes to AD pathology through these pathways. These findings highlight the significant role of lipid metabolism in neurodegeneration and offer potential therapeutic targets for age-related diseases, including AD.
Collapse
Affiliation(s)
- Gwangho Yoon
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Min Kyoung Kam
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Young Ho Koh
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| | - Chulman Jo
- Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju-si, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
14
|
Shroff R, Goetzl EJ. Protein Kinases - High Yield Targets for Cancer and Dementia Drug Discovery. Am J Med 2024; 137:1055-1058. [PMID: 38925498 DOI: 10.1016/j.amjmed.2024.06.016] [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: 06/11/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
Cellular protein kinases are involved in diverse normal cellular functions. Many types of dysregulation of protein kinases are the molecular basis for development of common cancers and neurodegenerative diseases. More than 80 small-molecule protein kinase inhibitors are available now and approved by the US Food and Drug Administration for successful treatment of cancers and neurodegenerative diseases. Newly designed protein kinase inhibitors and related forms of therapy based on a greater understanding of molecular mechanisms have diminished the appearance of disease resistance to protein kinase inhibitors and other side effects. These advances will further promote the success of protein kinase inhibitors in treatment of common cancers, Alzheimer's disease, and other neurodegenerative conditions.
Collapse
Affiliation(s)
- Rachna Shroff
- Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Edward J Goetzl
- Department of Medicine, University of California Medical Center, San Francisco.
| |
Collapse
|
15
|
Martínez-Drudis L, Bérard M, Musiol D, Rivest S, Oueslati A. Pharmacological inhibition of PLK2 kinase activity mitigates cognitive decline but aggravates APP pathology in a sex-dependent manner in APP/PS1 mouse model of Alzheimer's disease. Heliyon 2024; 10:e39571. [PMID: 39498012 PMCID: PMC11532864 DOI: 10.1016/j.heliyon.2024.e39571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Converging evidence from clinical and experimental studies suggest the potential significance of Polo-like kinase 2 (PLK2) in regulating the phosphorylation and toxicity of the Alzheimer's disease (AD)-related protein, amyloid precursor protein (APP). These findings have prompted various experimental trials aimed at inhibiting PLK2 kinase activity in different transgenic mouse models of AD. While positive impacts on cognitive decline were reported in these studies, the cellular effects remained controversial. In the present study, we sought to assess the cognitive and cellular consequences of chronic PLK2 inhibitor treatment in the APP/PS1 transgenic mouse model of AD. First, we confirmed that inhibiting PLK2 prevented cognitive decline in a sex-dependent manner, particularly by enhancing working memory in male APP/PS1 mice. Surprisingly, cellular analysis revealed that treatment with PLK2 inhibitor increased the load of amyloid plaques and elevated levels of soluble amyloid β (Aβ) 40 and Aβ42 in the cortex, as well as insoluble Aβ42 in the hippocampus of female mice, without affecting APP pathology in males. These results underscore the potential of PLK2 inhibition to mitigate cognitive symptoms in males. However, paradoxically, it intensifies amyloid pathology in females by enhancing APP amyloidogenic processing, creating a controversial aspect to its therapeutic impact. Overall, these data highlight the sex-dependent nature of the effects of PLK2 inhibition, which may also be influenced by the genetic background of the transgenic mouse model utilized.
Collapse
Affiliation(s)
- Laura Martínez-Drudis
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Morgan Bérard
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Dylan Musiol
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Serge Rivest
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Abid Oueslati
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| |
Collapse
|
16
|
Feng G, Yang X, Shuai W, Wang G, Ouyang L. Update on JNK inhibitor patents: 2015 to present. Expert Opin Ther Pat 2024; 34:907-927. [PMID: 39223788 DOI: 10.1080/13543776.2024.2400167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION c-Jun N-terminal kinase (JNK) regulates various biological processes through the phosphorylation cascade and is closely associated with numerous diseases, including inflammation, cardiovascular diseases, and neurological disorders. Therefore, JNKs have emerged as potential targets for disease treatment. AREAS COVERED This review compiles the patents and literatures concerning JNK inhibitors through retrieving relevant information from the SciFinder, Google Patents databases, and PubMed from 2015 to the present. It summarizes the structure-activity relationship (SAR) and biological activity profiles of JNK inhibitors, offering valuable perspectives on their potential therapeutic applications. EXPERT OPINION The JNK kinase serves as a novel target for the treatment of neurodegenerative disorders, pulmonary fibrosis, and other illnesses. A variety of small-molecule inhibitors targeting JNKs have demonstrated promising therapeutic potential in preclinical studies, which act upon JNK kinases via distinct mechanisms, encompassing traditional ATP competitive inhibition, covalent inhibition, and bidentate inhibition. Among them, several JNK inhibitors from PregLem SA, Celegene SA, and Xigen SA have accomplished the early stage of clinical trials, and their results will guide the development and indications of future JNK inhibitors.
Collapse
Affiliation(s)
| | | | | | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and West China second Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Liang Ouyang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and West China second Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
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: 140] [Impact Index Per Article: 140.0] [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
|
18
|
Shareghi-Boroujeni D, Iraji A, Dara M, Hashempur MH, Zare S, Hariri R, Akbarzadeh T, Saeedi M. Synthesis of novel hybrids of 1,2,3-triazoles-hydrazone: targeting cholinesterases and Alzheimer's related genes. Future Med Chem 2024; 16:1519-1535. [PMID: 38864182 PMCID: PMC11370907 DOI: 10.1080/17568919.2024.2359894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/13/2024] [Indexed: 06/13/2024] Open
Abstract
Aim: A new series of 1,2,3-triazole-hydrazone derivatives were developed to evaluate their anti-Alzheimer's activity. Materials & methods: All compounds were screened toward cholinesterases via the modified Ellman's method. The toxicity assay on SH-SY5Y cells was performed using the MTT assay, and the expression levels of GSK-3α, GSK-3β, DYRK1 and CDK5 were assessed in the presence of compounds 6m and 6p.Results:6m and 6p; acting as mixed-type inhibitors, exhibited promising acetylcholinesterase and butyrylcholinesterase inhibitory activity, respectively. 6m demonstrated no toxicity under tested concentrations on the SH-SY5Y cells and positively impacted neurodegenerative pathways. Notably, 6m displayed a significant downregulation in mRNA levels of GSK-3α, GSK-3β and CDK5.Conclusion: The target compounds could be considered in developing anti-Alzheimer's disease agents.
Collapse
Affiliation(s)
- Diba Shareghi-Boroujeni
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Iraji
- Research Center for Traditional Medicine & History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahintaj Dara
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hashem Hashempur
- Research Center for Traditional Medicine & History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahrokh Zare
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roshanak Hariri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Akbarzadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Persian Medicine & Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Saeedi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Persian Medicine & Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Fan M, Song X, Lu L, He J, Shen Y, Zhang C, Wang F, Xie Y. Comprehensive safety evaluation of a novel multitargeting compound XYY-CP1106: A candidate for Alzheimer's disease. Biomed Pharmacother 2024; 176:116786. [PMID: 38805971 DOI: 10.1016/j.biopha.2024.116786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
Multitargeting has become a promising strategy for the development of anti-Alzheimer's disease (AD) drugs, considering the complexity of molecular mechanisms in AD pathology. In most pre-clinical studies, the effectiveness of these multi-targeted anti-AD drugs has been demonstrated but comprehensive safety assessments are lacking. Here, the safety evaluation of a novel multi-targeted candidate in AD (XYY-CP1106), characterized by its dual-property of iron chelation and monoamine oxidase B inhibition, was conducted by multifaceted analysis. Acute toxicity in mice was conducted to investigate the safety of oral administration and the maximum tolerated dose of the agent. In vitro Ames analysis, CHL chromosomal aberration analysis, and bone marrow micronucleus analysis were executed to evaluate the genotoxicity. A teratogenesis investigation in pregnant mice were meticulously performed to evaluate the teratogenesis of XYY-CP1106. Furthermore, a 90-day long-term toxicity analysis in rats was investigated to evaluate the cumulative toxicity after long-term administration. Strikingly, no toxic phenomena were found in all investigations, demonstrating relatively high safety profile of the candidate compound. The securing of safety heightened the translational significance of XYY-CP1106 as a novel multi-targeted anti-AD candidate, supporting the rationality of multitargeting strategy in the designs of smart anti-AD drugs.
Collapse
Affiliation(s)
- Miaoliang Fan
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiaoxin Song
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Liwen Lu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jiayan He
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yikai Shen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Changjun Zhang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Fang Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Yuanyuan Xie
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceutical, Zhejiang University of Technology, Hangzhou 310014, China; Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, China; Key Laboratory of Pharmaceutical Engineering of Zhejiang Province, China.
| |
Collapse
|
20
|
Zhang T, Kim BM, Lee TH. Death-associated protein kinase 1 as a therapeutic target for Alzheimer's disease. Transl Neurodegener 2024; 13:4. [PMID: 38195518 PMCID: PMC10775678 DOI: 10.1186/s40035-023-00395-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/18/2023] [Indexed: 01/11/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly and represents a major clinical challenge in the ageing society. Neuropathological hallmarks of AD include neurofibrillary tangles composed of hyperphosphorylated tau, senile plaques derived from the deposition of amyloid-β (Aβ) peptides, brain atrophy induced by neuronal loss, and synaptic dysfunctions. Death-associated protein kinase 1 (DAPK1) is ubiquitously expressed in the central nervous system. Dysregulation of DAPK1 has been shown to contribute to various neurological diseases including AD, ischemic stroke and Parkinson's disease (PD). We have established an upstream effect of DAPK1 on Aβ and tau pathologies and neuronal apoptosis through kinase-mediated protein phosphorylation, supporting a causal role of DAPK1 in the pathophysiology of AD. In this review, we summarize current knowledge about how DAPK1 is involved in various AD pathological changes including tau hyperphosphorylation, Aβ deposition, neuronal cell death and synaptic degeneration. The underlying molecular mechanisms of DAPK1 dysregulation in AD are discussed. We also review the recent progress regarding the development of novel DAPK1 modulators and their potential applications in AD intervention. These findings substantiate DAPK1 as a novel therapeutic target for the development of multifunctional disease-modifying treatments for AD and other neurological disorders.
Collapse
Affiliation(s)
- Tao Zhang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, Fujian, China
| | - Byeong Mo Kim
- Research Center for New Drug Development, AgingTarget Inc., 10F Ace Cheonggye Tower, 53, Seonggogae-Ro, Uiwang-Si, 16006, Gyeonggi-Do, Korea.
| | - Tae Ho Lee
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
21
|
Meijer L, Chrétien E, Ravel D. Leucettinib-21, a DYRK1A Kinase Inhibitor as Clinical Drug Candidate for Alzheimer's Disease and Down Syndrome. J Alzheimers Dis 2024; 101:S95-S113. [PMID: 39422950 DOI: 10.3233/jad-240078] [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: 10/19/2024]
Abstract
Alzheimer's disease (AD) and Down syndrome (DS) share a common therapeutic target, the dual-specificity, tyrosine phosphorylation activated kinase 1A (DYRK1A). Abnormally active DYRK1A is responsible for cognitive disorders (memory, learning, spatial localization) observed in both conditions. In DS, DYRK1A is overexpressed due to the presence of the DYRK1A gene on chromosome 21. In AD, calcium-activated calpains cleave full-length DYRK1A (FL-DYRK1A) into a more stable and more active, low molecular weight, kinase (LMW-DYRK1A). Genetic and pharmacological experiments carried out with animal models of AD and DS strongly support the idea that pharmacological inhibitors of DYRK1A might be able to correct memory/learning disorders in people with AD and DS. Starting from a marine sponge natural product, Leucettamine B, Perha Pharmaceuticals has optimized, through classical medicinal chemistry, and extensively characterized a small molecule drug candidate, Leucettinib-21. Regulatory preclinical safety studies in rats and minipigs have been completed and formulation of Leucettinib-21 has been optimized as immediate-release tablets. Leucettinib-21 is now undergoing a phase 1 clinical trial (120 participants, including 12 adults with DS and 12 patients with AD). The therapeutic potential of DYRK1A inhibitors in AD and DS is presented.
Collapse
Affiliation(s)
- Laurent Meijer
- Perha Pharmaceuticals, Hôtel de Recherche, Roscoff, Bretagne, France
| | - Emilie Chrétien
- Perha Pharmaceuticals, Hôtel de Recherche, Roscoff, Bretagne, France
| | | |
Collapse
|
22
|
Fonódi M, Thalwieser Z, Csortos C, Boratkó A. TIMAP, a Regulatory Subunit of Protein Phosphatase 1, Inhibits In Vitro Neuronal Differentiation. Int J Mol Sci 2023; 24:17360. [PMID: 38139189 PMCID: PMC10744335 DOI: 10.3390/ijms242417360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
TIMAP (TGF-β-inhibited membrane associated protein) is abundant in endothelial cells, and it has been regarded as a member of the myosin phosphatase targeting protein (MYPT) family. Our workgroup previously identified several interacting protein partners of TIMAP and proved its regulatory subunit role for protein phosphatase 1 catalytic subunit (PP1c). TIMAP is also expressed in neuronal cells, but details of its function have not been studied yet. Therefore, we aimed to explore the role of TIMAP in neuronal cells, especially during differentiation. Expression of TIMAP was proved both at mRNA and protein levels in SH-SY5Y human neuroblastoma cells. Differentiation of SH-SY5Y cells was optimized and proved by the detection of neuronal differentiation markers, such as β3-tubulin, nestin and inhibitor of differentiation 1 (ID1) using qPCR and Western blot. We found downregulation of TIMAP during differentiation. In accordance with this, overexpression of recombinant TIMAP attenuated the differentiation of neuronal cells. Moreover, the subcellular localization of TIMAP has changed during differentiation as it translocated from the plasma membrane into the nucleus. The nuclear interactome of TIMAP revealed more than 50 proteins, offering the possibility to further investigate the role of TIMAP in several key physiological pathways of neuronal cells.
Collapse
Affiliation(s)
| | | | | | - Anita Boratkó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary; (M.F.); (Z.T.); (C.C.)
| |
Collapse
|