1
|
Sharma P, Sharma S, Yadav Y, Shukla P, Sagar R. Current pharmacophore based approaches for the development of new anti-Alzheimer's agents. Bioorg Med Chem 2024; 113:117926. [PMID: 39306973 DOI: 10.1016/j.bmc.2024.117926] [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: 07/24/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 10/13/2024]
Abstract
Amyloid beta peptide (Aβ) and hyperphosphorylated neuronal tau proteins accumulate in neurofibrillary tangles in Alzheimer's disease (AD), a chronic neurodegenerative illness. Chronic inflammation in the brain, which promotes disease progression, is another feature of the Alzheimer's disease pathogenesis. Approximately 60-70 % of dementia cases are caused by AD. The development of effective therapies for the treatment of AD is urgently needed given the severity of the condition and its rapidly rising prevalence. Cholinesterase inhibitors, beta-amyloid (A-beta), tau inhibitors, and many other medications are currently used as preventive medicine for AD. These medications can temporarily suppress dementia symptoms but cannot halt or reverse the disease's progression. Many international pharmaceutical companies have tried numerous times to develop an amyloid clearing medication based on the amyloid hypothesis, but without success. Therefore, the amyloid theory may not be entirely plausible. This review mainly covers the recent and important reported pharmacophores as the starting point to discuss already known targets like tau, butyrylcholinesterase, amyloid beta, and acetylcholinesterase and covers the literature between years 2019-2024.
Collapse
Affiliation(s)
- Prachi Sharma
- Department of Chemistry, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Sunil Sharma
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Yogesh Yadav
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Paritosh Shukla
- Department of Chemistry, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India.
| | - Ram Sagar
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
2
|
Shen Y, Zhu W, Li S, Zhang Z, Zhang J, Li M, Zheng W, Wang D, Zhong Y, Li M, Zheng H, Du J. Integrated analyses of 5 mC, 5hmC methylation and gene expression reveal pathology-associated AKT3 gene and potential biomarkers for Alzheimer's disease. J Psychiatr Res 2024; 178:367-377. [PMID: 39197298 DOI: 10.1016/j.jpsychires.2024.08.021] [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: 03/22/2024] [Revised: 07/18/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
AIMS 5 mC methylation and hydroxymethylation (5hmC) are associated with Alzheimer's disease (AD). However, previous studies were limited by the absence of a 5hmC calculation. This study aims to find AD associated predictors and potential therapeutic chemicals using bioinformatics approach integrating 5 mC, 5hmC, and expression changes, and an AD mouse model. METHODS Gene expression microarray and 5 mC and 5hmC sequencing datasets were downloaded from GEO repository. 142 AD and 52 normal entorhinal cortex specimens were enrolled. Data from oxidative bisulfite sequencing (oxBS)-treated samples, which represent only 5 mC, were used to calculate 5hmC level. Functional analyses, random forest supervised classification and methylation validation were applied. Potential chemicals were predicted by CMap. Morris water maze, Y maze and novel object recognition behavior tests were performed using FAD4T AD mice model. Cortex and hippocampus tissues were isolated for immunohistochemical staining. RESULTS C1QTNF5, UBD, ZFP106, NEDD1, AKT3, and MBP genes involving 13 promoter CpG sites with 5mc, 5hmC methylation and expression difference were identified. AKT3 and MBP were down-regulated in both patients and mouse model. Three CpG sites in AKT3 and MBP showed significant methylation difference on validation. FAD4T AD mice showed recession in brain functions and lower AKT3 expression in both cortex and hippocampus. Ten chemicals were predicted as potential treatments for AD. CONCLUSIONS AKT3 and MBP may be associated with AD pathology and could serve as biomarkers. The ten predicted chemicals might offer new therapeutic approaches. Our findings could contribute to identifying novel markers and advancing the understanding of AD mechanisms.
Collapse
Affiliation(s)
- Yupei Shen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Weiqiang Zhu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Shuaicheng Li
- School of Computer Science, Fudan University, Shanghai Key Laboratory of Intelligent Information Processing Shanghai, China
| | - Zhaofeng Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jian Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Mingjie Li
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Wei Zheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Difei Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yushun Zhong
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Min Li
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Huajun Zheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jing Du
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.
| |
Collapse
|
3
|
Putra M, Rao NS, Gardner C, Liu G, Trommater J, Bunney M, Gage M, Bassuk AG, Hefti M, Lee G, Thippeswamy T. Enhanced Fyn-tau and NR2B-PSD95 interactions in epileptic foci in experimental models and human epilepsy. Brain Commun 2024; 6:fcae327. [PMID: 39355003 PMCID: PMC11444080 DOI: 10.1093/braincomms/fcae327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 10/03/2024] Open
Abstract
Epilepsy and Alzheimer's disease share some common pathologies such as neurodegeneration, seizures and impaired cognition. However, the molecular mechanisms of these changes are still largely unknown. Fyn, a Src-family non-receptor tyrosine kinase (SFK), and its interaction with tau in mediating brain pathology in epilepsy and Alzheimer's disease can be a potential therapeutic target for disease modification. Although Fyn and tau pathology occurs in both Alzheimer's disease and epilepsy, the dynamics of Fyn-tau and PSD95-NR2B interactions affected by seizures and their impact on brain pathology in epilepsy have not been investigated. In this study, we demonstrate a significant increase of Fyn-tau interactions following seizure induction by kainate in both acute and chronic rodent models and in human epilepsy. In the early phase of epileptogenesis, we show increased Fyn/tau/NR2B/PSD95/neuronal nitric oxide synthase complexes after status epilepticus and a postsynaptic increase of phosphorylated tau (pY18 and AT8), Fyn (pSFK-Y416), NMDAR (pNR2B-Y1472) and neuronal nitric oxide synthase. Hippocampal proximity ligation assay and co-immunoprecipitation revealed a sustained increase of Fyn-tau and NR2B-PSD95 complexes/binding in rat chronic epilepsy at 3 months post-status epilepticus. Enhanced Fyn-tau complexes strongly correlated with the frequency of spontaneously recurring convulsive seizures and epileptiform spikes in the chronic epilepsy model. In human epileptic brains, we also identified increased Fyn-tau and NR2B-PSD95 complexes, tau phosphorylation (pY18 and AT8) and Fyn activation (pSFK-Y416), implying the translational and therapeutic potential of these molecular interactions. In tau knockout mice and in rats treated with a Fyn/SFK inhibitor saracatinib, we found a significant reduction of phosphorylated Fyn, tau (AT8 in saracatinib-treated), NR2B and neuronal nitric oxide synthase and their interactions (Fyn-tau and NR2B-PSD95 in saracatinib-treated group; NR2B-PSD95 in tau knockout group). The reduction of Fyn-tau and NR2B-PSD95 interactions in the saracatinib-treated group, in contrast to the vehicle-treated group, correlated with the modification in seizure progression in the rat chronic epilepsy model. These findings from animal models and human epilepsy provide evidence for the role of Fyn-tau and NR2B-PSD95 interactions in seizure-induced brain pathology and suggest that blocking such interactions could modify the progression of epilepsy.
Collapse
Affiliation(s)
- Marson Putra
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Nikhil S Rao
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Cara Gardner
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Guanghao Liu
- Department of Internal Medicine, Carver College of Medicine, Carver College of Medicine University of Iowa, Iowa City, IA 52242, USA
| | - Jordan Trommater
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Michael Bunney
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Meghan Gage
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Alexander G Bassuk
- Department of Pediatrics, The University of Iowa Stead Family, Iowa City, IA 52242, USA
- Department of Neurology, The University of Iowa Stead Family, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute (INI), College of Medicine, University of Iowa Carver, Iowa City, IA 52242, USA
| | - Marco Hefti
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52240, USA
| | - Gloria Lee
- Department of Internal Medicine, Carver College of Medicine, Carver College of Medicine University of Iowa, Iowa City, IA 52242, USA
| | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| |
Collapse
|
4
|
Cruz E, Nisbet RM, Götz J. Break and accelerator-The mechanics of Tau (and amyloid) toxicity. Cytoskeleton (Hoboken) 2024; 81:24-29. [PMID: 37632370 DOI: 10.1002/cm.21781] [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: 06/28/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023]
Abstract
Aggregates of the microtubule-associated protein Tau define more than a dozen primary tauopathies, and together with amyloid-β, the secondary tauopathy Alzheimer's disease (AD). Historically, Tau has been viewed as executor of amyloid-β toxicity, with the two molecules working together as "trigger and bullet." Given the two protein's opposing roles in protein translation, we wish to introduce another metaphor, borrowing from the mechanics of a car, with amyloid-β boosting Tau translation, whereas Tau puts a break on global translation. The underlying studies entail an alternative hypothesis regarding Tau's subcellular accumulation in AD, namely its de novo synthesis in the somatodendritic domain rather than the relocalization from the axon upon dissociation from microtubules. We contest that it may be worth (given Tau's 50th birthday) to revisit some entrenched dogmas about Tau's pathophysiology.
Collapse
Affiliation(s)
- Esteban Cruz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Rebecca M Nisbet
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
5
|
Leinenga G, Padmanabhan P, Götz J. Improving Cognition Without Clearing Amyloid: Effects of Tau and Ultrasound Neuromodulation. J Alzheimers Dis 2024; 100:S211-S222. [PMID: 39058447 DOI: 10.3233/jad-240616] [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: 07/28/2024]
Abstract
Alzheimer's disease is characterized by progressive impairment of neuronal functions culminating in neuronal loss and dementia. A universal feature of dementia is protein aggregation, a process by which a monomer forms intermediate oligomeric assembly states and filaments that develop into end-stage hallmark lesions. In Alzheimer's disease, this is exemplified by extracellular amyloid-β (Aβ) plaques which have been placed upstream of tau, found in intracellular neurofibrillary tangles and dystrophic neurites. This implies causality that can be modeled as a linear activation cascade. When Aβ load is reduced, for example, in response to an anti-Aβ immunotherapy, cognitive functions improve in plaque-forming mice. They also deteriorate less in clinical trial cohorts although real-world clinical benefits remain to be demonstrated. Given the existence of aged humans with unimpaired cognition despite a high plaque load, the central role of Aβ has been challenged. A counter argument has been that clinical symptoms would eventually develop if these aged individuals were to live long enough. Alternatively, intrinsic mechanisms that protect the brain in the presence of pathology may exist. In fact, Aβ toxicity can be abolished by either reducing or manipulating tau (through which Aβ signals), at least in preclinical models. In addition to manipulating steps in this linear pathocascade model, mechanisms of restoring brain reserve can also counteract Aβ toxicity. Low-intensity ultrasound is a neuromodulatory modality that can improve cognitive functions in Aβ-depositing mice without the need for removing Aβ. Together, this highlights a dissociation of Aβ and cognition, with important implications for therapeutic interventions.
Collapse
Affiliation(s)
- Gerhard Leinenga
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
| | - Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
6
|
Ali T, Klein AN, Vu A, Arifin MI, Hannaoui S, Gilch S. Peptide aptamer targeting Aβ-PrP-Fyn axis reduces Alzheimer's disease pathologies in 5XFAD transgenic mouse model. Cell Mol Life Sci 2023; 80:139. [PMID: 37149826 PMCID: PMC10164677 DOI: 10.1007/s00018-023-04785-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 04/11/2023] [Accepted: 04/22/2023] [Indexed: 05/08/2023]
Abstract
Currently, no effective therapeutics exist for the treatment of incurable neurodegenerative diseases such as Alzheimer's disease (AD). The cellular prion protein (PrPC) acts as a high-affinity receptor for amyloid beta oligomers (AβO), a main neurotoxic species mediating AD pathology. The interaction of AβO with PrPC subsequently activates Fyn tyrosine kinase and neuroinflammation. Herein, we used our previously developed peptide aptamer 8 (PA8) binding to PrPC as a therapeutic to target the AβO-PrP-Fyn axis and prevent its associated pathologies. Our in vitro results indicated that PA8 prevents the binding of AβO with PrPC and reduces AβO-induced neurotoxicity in mouse neuroblastoma N2a cells and primary hippocampal neurons. Next, we performed in vivo experiments using the transgenic 5XFAD mouse model of AD. The 5XFAD mice were treated with PA8 and its scaffold protein thioredoxin A (Trx) at a 14.4 µg/day dosage for 12 weeks by intraventricular infusion through Alzet® osmotic pumps. We observed that treatment with PA8 improves learning and memory functions of 5XFAD mice as compared to Trx-treated 5XFAD mice. We found that PA8 treatment significantly reduces AβO levels and Aβ plaques in the brain tissue of 5XFAD mice. Interestingly, PA8 significantly reduces AβO-PrP interaction and its downstream signaling such as phosphorylation of Fyn kinase, reactive gliosis as well as apoptotic neurodegeneration in the 5XFAD mice compared to Trx-treated 5XFAD mice. Collectively, our results demonstrate that treatment with PA8 targeting the AβO-PrP-Fyn axis is a promising and novel approach to prevent and treat AD.
Collapse
Affiliation(s)
- Tahir Ali
- Calgary Prion Research Unit, Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Antonia N Klein
- Calgary Prion Research Unit, Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Alex Vu
- Calgary Prion Research Unit, Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Maria I Arifin
- Calgary Prion Research Unit, Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Samia Hannaoui
- Calgary Prion Research Unit, Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Sabine Gilch
- Calgary Prion Research Unit, Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada.
- Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
7
|
Khan KM, Balasubramanian N, Gaudencio G, Wang R, Selvakumar GP, Kolling L, Pierson S, Tadinada SM, Abel T, Hefti M, Marcinkiewcz CA. Human tau-overexpressing mice recapitulate brainstem involvement and neuropsychiatric features of early Alzheimer's disease. Acta Neuropathol Commun 2023; 11:57. [PMID: 37009893 PMCID: PMC10069039 DOI: 10.1186/s40478-023-01546-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/07/2023] [Indexed: 04/04/2023] Open
Abstract
Alzheimer's disease (AD) poses an ever-increasing public health concern as the population ages, affecting more than 6 million Americans. AD patients present with mood and sleep changes in the prodromal stages that may be partly driven by loss of monoaminergic neurons in the brainstem, but a causal relationship has not been firmly established. This is due in part to a dearth of animal models that recapitulate early AD neuropathology and symptoms. The goal of the present study was to evaluate depressive and anxiety-like behaviors in a mouse model of AD that overexpresses human wild-type tau (htau) prior to the onset of cognitive impairments and assess these behavior changes in relationship to tau pathology, neuroinflammation, and monoaminergic dysregulation in the dorsal raphe nucleus (DRN) and locus coeruleus (LC). We observed depressive-like behaviors at 4 months in both sexes and hyperlocomotion in male htau mice. Deficits in social interaction persisted at 6 months and were accompanied by an increase in anxiety-like behavior in males. The behavioral changes at 4 months coincided with a lower density of serotonergic (5-HT) neurons, downregulation of 5-HT markers, reduced excitability of 5-HT neurons, and hyperphosphorylated tau in the DRN. Inflammatory markers were also upregulated in the DRN along with protein kinases and transglutaminase 2, which may promote tau phosphorylation and aggregation. Loss of 5-HT innervation to the entorhinal cortex and dentate gyrus of the hippocampus was also observed and may have contributed to depressive-like behaviors. There was also reduced expression of noradrenergic markers in the LC along with elevated phospho-tau expression, but this did not translate to a functional change in neuronal excitability. In total, these results suggest that tau pathology in brainstem monoaminergic nuclei and the resulting loss of serotonergic and/or noradrenergic drive may underpin depressive- and anxiety-like behaviors in the early stages of AD.
Collapse
Affiliation(s)
- Kanza M Khan
- Department of Neuroscience and Pharmacology, University of Iowa, 2-430 Bowen Science Building, Iowa City, IA, 52242, USA
- Psychological Sciences Department, Daemen University, Amherst, NY, 14226, USA
| | - Nagalakshmi Balasubramanian
- Department of Neuroscience and Pharmacology, University of Iowa, 2-430 Bowen Science Building, Iowa City, IA, 52242, USA
| | - Gabriel Gaudencio
- Department of Neuroscience and Pharmacology, University of Iowa, 2-430 Bowen Science Building, Iowa City, IA, 52242, USA
| | - Ruixiang Wang
- Department of Neuroscience and Pharmacology, University of Iowa, 2-430 Bowen Science Building, Iowa City, IA, 52242, USA
| | | | - Louis Kolling
- Department of Neuroscience and Pharmacology, University of Iowa, 2-430 Bowen Science Building, Iowa City, IA, 52242, USA
| | - Samantha Pierson
- Department of Neuroscience and Pharmacology, University of Iowa, 2-430 Bowen Science Building, Iowa City, IA, 52242, USA
| | - Satya M Tadinada
- Department of Neuroscience and Pharmacology, University of Iowa, 2-430 Bowen Science Building, Iowa City, IA, 52242, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, University of Iowa, 2-430 Bowen Science Building, Iowa City, IA, 52242, USA
| | - Marco Hefti
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Catherine A Marcinkiewcz
- Department of Neuroscience and Pharmacology, University of Iowa, 2-430 Bowen Science Building, Iowa City, IA, 52242, USA.
| |
Collapse
|
8
|
Liu X, Lai LY, Chen JX, Li X, Wang N, Zhou LJ, Jiang XW, Hu XL, Liu WW, Jiao XM, Qi ZT, Liu WJ, Wu LM, Huang YG, Xu ZH, Zhao QC. An inhibitor with GSK3β and DYRK1A dual inhibitory properties reduces Tau hyperphosphorylation and ameliorates disease in models of Alzheimer's disease. Neuropharmacology 2023; 232:109525. [PMID: 37004752 DOI: 10.1016/j.neuropharm.2023.109525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023]
Abstract
Since Alzheimer's disease (AD) is a complex and multifactorial neuropathology, the discovery of multi-targeted inhibitors has gradually demonstrated greater therapeutic potential. Neurofibrillary tangles (NFTs), the main neuropathologic hallmarks of AD, are mainly associated with hyperphosphorylation of the microtubule-associated protein Tau. The overexpression of GSK3β and DYRK1A has been recognized as an important contributor to hyperphosphorylation of Tau, leading to the strategy of using dual-targets inhibitors for the treatment of this disorder. ZDWX-12 and ZDWX-25, as harmine derivatives, were found good inhibition on dual targets in our previous study. Here, we firstly evaluated the inhibition effect of Tau hyperphosphorylation using two compounds by HEK293-Tau P301L cell-based model and okadaic acid (OKA)-induced mouse model. We found that ZDWX-25 was more effective than ZDWX-12. Then, based on comprehensively investigations on ZDWX-25 in vitro and in vivo, 1) the capability of ZDWX-25 to show a reduction in phosphorylation of multiple Tau epitopes in OKA-induced neurodegeneration cell models, and 2) the effect of reduction on NFTs by 3xTg-AD mouse model under administration of ZDWX-25, an orally bioavailable, brain-penetrant dual-targets inhibitor with low toxicity. Our data highlight that ZDWX-25 is a promising drug for treating AD.
Collapse
|
9
|
Fyn nanoclustering requires switching to an open conformation and is enhanced by FTLD-Tau biomolecular condensates. Mol Psychiatry 2023; 28:946-962. [PMID: 36258016 PMCID: PMC9908554 DOI: 10.1038/s41380-022-01825-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/08/2022]
Abstract
Fyn is a Src kinase that controls critical signalling cascades and has been implicated in learning and memory. Postsynaptic enrichment of Fyn underpins synaptotoxicity in dementias such as Alzheimer's disease and frontotemporal lobar degeneration with Tau pathology (FTLD-Tau). The FLTD P301L mutant Tau is associated with a higher propensity to undergo liquid-liquid phase separation (LLPS) and form biomolecular condensates. Expression of P301L mutant Tau promotes aberrant trapping of Fyn in nanoclusters within hippocampal dendrites by an unknown mechanism. Here, we used single-particle tracking photoactivated localisation microscopy to demonstrate that the opening of Fyn into its primed conformation promotes its nanoclustering in dendrites leading to increased Fyn/ERK/S6 downstream signalling. Preventing the auto-inhibitory closed conformation of Fyn through phospho-inhibition or through perturbation of its SH3 domain increased Fyn's nanoscale trapping, whereas inhibition of the catalytic domain had no impact. By combining pharmacological and genetic approaches, we demonstrate that P301L Tau enhanced both Fyn nanoclustering and Fyn/ERK/S6 signalling via its ability to form biomolecular condensates. Together, our findings demonstrate that Fyn alternates between a closed and an open conformation, the latter being enzymatically active and clustered. Furthermore, pathogenic immobilisation of Fyn relies on the ability of P301L Tau to form biomolecular condensates, thus highlighting the critical importance of LLPS in controlling nanoclustering and downstream intracellular signalling events.
Collapse
|
10
|
Liu YS, Zhao HF, Li Q, Cui HW, Huang GD. Research Progress on the Etiology and Pathogenesis of Alzheimer's Disease from the Perspective of Chronic Stress. Aging Dis 2022:AD.2022.1211. [PMID: 37163426 PMCID: PMC10389837 DOI: 10.14336/ad.2022.1211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/12/2022] [Indexed: 05/12/2023] Open
Abstract
Due to its extremely complex pathogenesis, no effective drugs to prevent, delay progression, or cure Alzheimer's disease (AD) exist at present. The main pathological features of AD are senile plaques composed of β-amyloid, neurofibrillary tangles formed by hyperphosphorylation of the tau protein, and degeneration or loss of neurons in the brain. Many risk factors associated with the onset of AD, including gene mutations, aging, traumatic brain injury, endocrine and cardiovascular diseases, education level, and obesity. Growing evidence points to chronic stress as one of the major risk factors for AD, as it can promote the onset and development of AD-related pathologies via a mechanism that is not well known. The use of murine stress models, including restraint, social isolation, noise, and unpredictable stress, has contributed to improving our understanding of the relationship between chronic stress and AD. This review summarizes the evidence derived from murine models on the pathological features associated with AD and the related molecular mechanisms induced by chronic stress. These results not only provide a retrospective interpretation for understanding the pathogenesis of AD, but also provide a window of opportunity for more effective preventive and identifying therapeutic strategies for stress-induced AD.
Collapse
Affiliation(s)
- Yun-Sheng Liu
- Department of Neurosurgery, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Hua-Fu Zhao
- Department of Neurosurgery, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Qian Li
- Department of Neurosurgery, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Han-Wei Cui
- The Central Laboratory, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Central Laboratory, Shenzhen Samii Medical Center, Shenzhen, China
| | - Guo-Dong Huang
- Department of Neurosurgery, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
11
|
Fortner A, Chera A, Tanca A, Bucur O. Apoptosis regulation by the tyrosine-protein kinase CSK. Front Cell Dev Biol 2022; 10:1078180. [PMID: 36578781 PMCID: PMC9792154 DOI: 10.3389/fcell.2022.1078180] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
C-terminal Src kinase (CSK) is a cytosolic tyrosine-protein kinase with an important role in regulating critical cellular decisions, such as cellular apoptosis, survival, proliferation, cytoskeletal organization and many others. Current knowledge on the CSK mechanisms of action, regulation and functions is still at an early stage, most of CSK's known actions and functions being mediated by the negative regulation of the SRC family of tyrosine kinases (SFKs) through phosphorylation. As SFKs play a vital role in apoptosis, cell proliferation and survival regulation, SFK inhibition by CSK has a pro-apoptotic effect, which is mediated by the inhibition of cellular signaling cascades controlled by SFKs, such as the MAPK/ERK, STAT3 and PI3K/AKT signaling pathways. Abnormal functioning of CSK and SFK activation can lead to diseases such as cancer, cardiovascular and neurological manifestations. This review describes apoptosis regulation by CSK, CSK inhibition of the SFKs and further explores the clinical relevance of CSK in important pathologies, such as cancer, autoimmune, autoinflammatory, neurologic diseases, hypertension and HIV/AIDS.
Collapse
Affiliation(s)
- Andra Fortner
- Victor Babes National Institute of Pathology, Bucharest, Romania,Medical School, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - Alexandra Chera
- Victor Babes National Institute of Pathology, Bucharest, Romania,Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Antoanela Tanca
- Victor Babes National Institute of Pathology, Bucharest, Romania,Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania,*Correspondence: Octavian Bucur, ; Antoanela Tanca,
| | - Octavian Bucur
- Victor Babes National Institute of Pathology, Bucharest, Romania,Viron Molecular Medicine Institute, Boston, MA, United States,*Correspondence: Octavian Bucur, ; Antoanela Tanca,
| |
Collapse
|
12
|
Garrido A, Santamaría E, Fernández-Irigoyen J, Soto M, Simonet C, Fernández M, Obiang D, Tolosa E, Martí MJ, Padmanabhan S, Malagelada C, Ezquerra M, Fernández-Santiago R. Differential Phospho-Signatures in Blood Cells Identify LRRK2 G2019S Carriers in Parkinson's Disease. Mov Disord 2022; 37:1004-1015. [PMID: 35049090 PMCID: PMC9306798 DOI: 10.1002/mds.28927] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/14/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Background The clinicopathological phenotype of G2019S LRRK2‐associated Parkinson's disease (L2PD) is similar to idiopathic Parkinson's disease (iPD), and G2019S LRRK2 nonmanifesting carriers (L2NMCs) are at increased risk for development of PD. With various therapeutic strategies in the clinical and preclinical pipeline, there is an urgent need to identify biomarkers that can aid early diagnosis and patient enrichment for ongoing and future LRRK2‐targeted trials. Objective The objective of this work was to investigate differential protein and phospho‐protein changes related to G2019S mutant LRRK2 in peripheral blood mononuclear cells from G2019S L2PD patients and G2019S L2NMCs, identify specific phospho‐protein changes associated with the G2019S mutation and with disease status, and compare findings with patients with iPD. Methods We performed an unbiased phospho‐proteomic study by isobaric label–based mass spectrometry using peripheral blood mononuclear cell group pools from a LRRK2 cohort from Spain encompassing patients with G2019S L2PD (n = 20), G2019S L2NMCs (n = 20), healthy control subjects (n = 30), patients with iPD (n = 15), patients with R1441G L2PD (n = 5), and R1441G L2NMCs (n = 3) (total N = 93). Results Comparing G2019S carriers with healthy controls, we identified phospho‐protein changes associated with the G2019S mutation. Moreover, we uncovered a specific G2019S phospho‐signature that changes with disease status and can discriminate patients with G2019S L2PD, G2019S L2NMCs, and healthy controls. Although patients with iPD showed a differential phospho‐proteomic profile, biological enrichment analyses revealed similar changes in deregulated pathways across the three groups. Conclusions We found a differential phospho‐signature associated with LRRK2 G2019S for which, consistent with disease status, the phospho‐profile from PD at‐risk G2019S L2NMCs was more similar to healthy controls than patients with G2019S L2PD with the manifested disease. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society
Collapse
Affiliation(s)
- Alicia Garrido
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Enrique Santamaría
- Proteored-ISCIII, Proteomics Platform, Clinical Neuroproteomics Unit, Navarrabiomed, Departamento de Salud, UPNA, IdiSNA, Pamplona, Navarra, Spain
| | - Joaquín Fernández-Irigoyen
- Proteored-ISCIII, Proteomics Platform, Clinical Neuroproteomics Unit, Navarrabiomed, Departamento de Salud, UPNA, IdiSNA, Pamplona, Navarra, Spain
| | - Marta Soto
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Cristina Simonet
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Manel Fernández
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain.,Parkinson's Disease and Movement Disorders Group of the Institut de Neurociències (Universitat de Barcelona), Barcelona, Catalonia, Spain
| | - Donina Obiang
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Eduardo Tolosa
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - María-José Martí
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Shalini Padmanabhan
- The Michael J. Fox Foundation for Parkinson's Research, Grand Central Station, New York, New York, USA
| | - Cristina Malagelada
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain.,Department of Biomedicine, Faculty of Medicine, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Mario Ezquerra
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain
| | - Rubén Fernández-Santiago
- Parkinson Disease and Movement Disorders Unit, Neurology Service, Institut Clínic de Neurociències, Hospital Clínic de Barcelona, Barcelona, Catalonia, Spain.,Lab of Parkinson Disease & Other Neurodegenerative Movement Disorders, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Catalonia, Spain.,Histology Unit, Department of Biomedicine, Faculty of Medicine, Universitat de Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
13
|
Merelli A, Repetto M, Lazarowski A, Auzmendi J. Hypoxia, Oxidative Stress, and Inflammation: Three Faces of Neurodegenerative Diseases. J Alzheimers Dis 2021; 82:S109-S126. [PMID: 33325385 DOI: 10.3233/jad-201074] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cerebral hypoxia-ischemia can induce a wide spectrum of biologic responses that include depolarization, excitotoxicity, oxidative stress, inflammation, and apoptosis, and result in neurodegeneration. Several adaptive and survival endogenous mechanisms can also be activated giving an opportunity for the affected cells to remain alive, waiting for helper signals that avoid apoptosis. These signals appear to help cells, depending on intensity, chronicity, and proximity to the central hypoxic area of the affected tissue. These mechanisms are present not only in a large list of brain pathologies affecting commonly older individuals, but also in other pathologies such as refractory epilepsies, encephalopathies, or brain trauma, where neurodegenerative features such as cognitive and/or motor deficits sequelae can be developed. The hypoxia inducible factor 1α (HIF-1α) is a master transcription factor driving a wide spectrum cellular response. HIF-1α may induce erythropoietin (EPO) receptor overexpression, which provides the therapeutic opportunity to administer pharmacological doses of EPO to rescue and/or repair affected brain tissue. Intranasal administration of EPO combined with other antioxidant and anti-inflammatory compounds could become an effective therapeutic alternative, to avoid and/or slow down neurodegenerative deterioration without producing adverse peripheral effects.
Collapse
Affiliation(s)
- Amalia Merelli
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina
| | - Marisa Repetto
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Analítica y Fisicoquímica, Cátedra de Química General e Inorgánica; Instituto de Bioquímica y Medicina Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas (IBIMOL, UBA-CONICET), Argentina
| | - Alberto Lazarowski
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina
| | - Jerónimo Auzmendi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioqummica, Departamento de Bioquímica Clínica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| |
Collapse
|
14
|
Spreading of Alzheimer tau seeds is enhanced by aging and template matching with limited impact of amyloid-β. J Biol Chem 2021; 297:101159. [PMID: 34480901 PMCID: PMC8477193 DOI: 10.1016/j.jbc.2021.101159] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
In Alzheimer's disease (AD), deposition of pathological tau and amyloid-β (Aβ) drive synaptic loss and cognitive decline. The injection of misfolded tau aggregates extracted from human AD brains drives templated spreading of tau pathology within WT mouse brain. Here, we assessed the impact of Aβ copathology, of deleting loci known to modify AD risk (Ptk2b, Grn, and Tmem106b) and of pharmacological intervention with an Fyn kinase inhibitor on tau spreading after injection of AD tau extracts. The density and spreading of tau inclusions triggered by human tau seed were unaltered in the hippocampus and cortex of APPswe/PSEN1ΔE9 transgenic and AppNL-F/NL-F knock-in mice. In mice with human tau sequence replacing mouse tau, template matching enhanced neuritic tau burden. Human AD brain tau-enriched preparations contained aggregated Aβ, and the Aβ coinjection caused a redistribution of Aβ aggregates in mutant AD model mice. The injection-induced Aβ phenotype was spatially distinct from tau accumulation and could be ameliorated by depleting Aβ from tau extracts. These data suggest that Aβ and tau pathologies propagate by largely independent mechanisms after their initial formation. Altering the activity of the Fyn and Pyk2 (Ptk2b) kinases involved in Aβ-oligomer–induced signaling, or deleting expression of the progranulin and TMEM106B lysosomal proteins, did not alter the somatic tau inclusion burden or spreading. However, mouse aging had a prominent effect to increase the accumulation of neuritic tau after injection of human AD tau seeds into WT mice. These studies refine our knowledge of factors capable of modulating tau spreading.
Collapse
|
15
|
Ettcheto M, Cano A, Sanchez-López E, Verdaguer E, Folch J, Auladell C, Camins A. Masitinib for the treatment of Alzheimer's disease. Neurodegener Dis Manag 2021; 11:263-276. [PMID: 34412534 DOI: 10.2217/nmt-2021-0019] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The actual standard treatment for mild-to-moderately severe Alzheimer's disease only attacks its symptoms. Masitinib is a potent and selective phenylaminothiazole-type tyrosine kinase inhibitor which is currently in Phase III studies for the treatment of Alzheimer's disease (AD) with the aim of modifying its evolution and with multiple pharmacological targets such as inhibition of mast cells activity, inhibition of microglia activation, modulation of Aβ and Tau protein signaling pathway and prevention of synaptic damage. Here, we review the preclinical and clinical studies that investigated the administration of masitinib treatment in monotherapy in AD. All research studies revealed positive effects concerning the cognitive functions in AD and generally with good safety and tolerability.
Collapse
Affiliation(s)
- Miren Ettcheto
- Department of Pharmacology, Toxicology & Therapeutic Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Spain.,Institut de Neurociències (UBNeuro), University of Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Amanda Cano
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Spain.,Research Center & Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades - International University of Catalunya (UIC), Barcelona, Spain
| | - Elena Sanchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Spain
| | - Ester Verdaguer
- Department of Cellular Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Spain
| | - Jaume Folch
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Unit of Biochemistry & Pharmacology, Faculty of Medicine & Health Sciences, University of Rovira i Virgili, Reus (Tarragona), Spain
| | - Carme Auladell
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Cellular Biology, Physiology & Immunology, Faculty of Biology, University of Barcelona, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology & Therapeutic Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Spain.,Institut de Neurociències (UBNeuro), University of Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
16
|
Low CYB, Lee JH, Lim FTW, Lee C, Ballard C, Francis PT, Lai MKP, Tan MGK. Isoform-specific upregulation of FynT kinase expression is associated with tauopathy and glial activation in Alzheimer's disease and Lewy body dementias. Brain Pathol 2021; 31:253-266. [PMID: 33128789 PMCID: PMC8017997 DOI: 10.1111/bpa.12917] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/26/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
Cumulative data suggest the involvement of Fyn tyrosine kinase in Alzheimer's disease (AD). Previously, our group has shown increased immunoreactivities of the FynT isoform in AD neocortex (with no change in the alternatively spliced FynB isoform) which associated with neurofibrillary degeneration and reactive astrogliosis. Since both the aforementioned neuropathological features are also variably found in Lewy Body dementias (LBD), we investigated potential perturbations of Fyn expression in the post-mortem neocortex of patients with AD, as well as those diagnosed as having one of the two main subgroups of LBD: Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB). We found selective upregulation of FynT expression in AD, PDD, and DLB which also correlated with cognitive impairment. Furthermore, increased FynT expression correlated with hallmark neuropathological lesions, soluble β-amyloid, and phosphorylated tau, as well as markers of microglia and astrocyte activation. In line with the human post-mortem studies, cortical FynT expression in aged mice transgenic for human P301S tau was upregulated and further correlated with accumulation of aggregated phosphorylated tau as well as with microglial and astrocytic markers. Our findings provide further evidence for the involvement of FynT in neurodegenerative dementias, likely via effects on tauopathy and neuroinflammation.
Collapse
Affiliation(s)
- Clara Y. B. Low
- Department of Clinical Translational ResearchSingapore General HospitalOutramSingapore
| | - Jasinda H. Lee
- Department of PharmacologyYong Loo Lin School of MedicineKent RidgeSingapore
| | - Frances T. W. Lim
- Department of Clinical Translational ResearchSingapore General HospitalOutramSingapore
| | - Chingli Lee
- Department of Clinical Translational ResearchSingapore General HospitalOutramSingapore
| | - Clive Ballard
- Institute for Health ResearchUniversity of Exeter Medical SchoolExeterUK
| | - Paul T. Francis
- Institute for Health ResearchUniversity of Exeter Medical SchoolExeterUK
- Wolfson Centre for Age‐Related DiseasesKing's College LondonLondonUK
| | - Mitchell K. P. Lai
- Department of PharmacologyYong Loo Lin School of MedicineKent RidgeSingapore
- Institute for Health ResearchUniversity of Exeter Medical SchoolExeterUK
- Wolfson Centre for Age‐Related DiseasesKing's College LondonLondonUK
| | - Michelle G. K. Tan
- Department of Clinical Translational ResearchSingapore General HospitalOutramSingapore
- Department of PharmacologyYong Loo Lin School of MedicineKent RidgeSingapore
| |
Collapse
|
17
|
Putra M, Puttachary S, Liu G, Lee G, Thippeswamy T. Fyn-tau Ablation Modifies PTZ-Induced Seizures and Post-seizure Hallmarks of Early Epileptogenesis. Front Cell Neurosci 2020; 14:592374. [PMID: 33363455 PMCID: PMC7752812 DOI: 10.3389/fncel.2020.592374] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/16/2020] [Indexed: 12/22/2022] Open
Abstract
Both Fyn and tau have been associated with neuronal hyperexcitability and neurotoxicity in many tauopathies, including Alzheimer's disease (AD). Individual genetic ablation of fyn or tau appears to be protective against aberrant excitatory neuronal activities in AD and epilepsy models. It is, however, still unknown whether ablation of both Fyn and tau can likely elicit more profound anti-seizure and neuroprotective effects. Here, we show the effects of genetic deletion of Fyn and/or tau on seizure severity in response to pentylenetetrazole (PTZ)-induced seizure in mouse models and neurobiological changes 24 h post-seizures. We used Fyn KO (fyn -/-), tau KO (tau -/-), double knockout (DKO) (fyn -/- / tau -/-), and wild-type (WT) mice of the same genetic background. Both tau KO and DKO showed a significant increase in latency to convulsive seizures and significantly decreased the severity of seizures post-PTZ. Although Fyn KO did not differ significantly from WT, in response to PTZ, Fyn KO still had 36 ± 8% seizure reduction and a 30% increase in seizure latency compared to WT. Surprisingly, in contrast to WT, Fyn KO mice showed higher mortality in <20 min of seizure induction; these mice had severe hydrocephalous. None of the tau -/- and DKO died during the study. In response to PTZ, all KO groups showed a significant reduction in neurodegeneration and gliosis, in contrast to WT, which showed increased neurodegeneration [especially, parvalbumin (PV)-GABAergic interneurons] and gliosis. DKO mice had the most reduced gliosis. Immunohistochemically, phospho-tau (AT8, pS199/S202), Fyn expression, as well as Fyn-tau interaction as measured by PLA increased in WT post-PTZ. Moreover, hippocampal Western blots revealed increased levels of AT8, tyrosine phospho-tau (pY18), and phosphorylated Src tyrosine family kinases (pSFK) in PTZ-treated WT, but not in KO, compared to respective controls. Furthermore, PV interneurons were protected from PTZ-induced seizure effects in all KO mice. The levels of inwardly rectifying potassium (Kir 4.1) channels were also downregulated in astrocytes in the WT post-PTZ, while its levels did not change in KO groups. Overall, our results demonstrated the role of Fyn and tau in seizures and their impact on the mediators of early epileptogenesis in PTZ model.
Collapse
Affiliation(s)
- Marson Putra
- Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Sreekanth Puttachary
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Guanghao Liu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Gloria Lee
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Thimmasettappa Thippeswamy
- Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
18
|
Soeda Y, Takashima A. New Insights Into Drug Discovery Targeting Tau Protein. Front Mol Neurosci 2020; 13:590896. [PMID: 33343298 PMCID: PMC7744460 DOI: 10.3389/fnmol.2020.590896] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
Microtubule-associated protein tau is characterized by the fact that it is an intrinsically disordered protein due to its lack of a stable conformation and high flexibility. Intracellular inclusions of fibrillar forms of tau with a β-sheet structure accumulate in the brain of patients with Alzheimer's disease and other tauopathies. Accordingly, detachment of tau from microtubules and transition of tau from a disordered state to an abnormally aggregated state are essential events preceding the onset of tau-related diseases. Many reports have shown that this transition is caused by post-translational modifications, including hyperphosphorylation and acetylation. The misfolded tau is self-assembled and forms a tau oligomer before the appearance of tau inclusions. Animal and pathological studies using human samples have demonstrated that tau oligomer formation contributes to neuronal loss. During the progression of tauopathies, tau seeds are released from cells and incorporated into other cells, leading to the propagation of pathological tau aggregation. Accumulating evidence suggests several potential approaches for blocking tau-mediated toxicity: (1) direct inhibition of pathological tau aggregation and (2) inhibition of tau post-translational modifications that occur prior to pathological tau aggregation, (3) inhibition of tau propagation and (4) stabilization of microtubules. In addition to traditional low-molecular-weight compounds, newer drug discovery approaches such as the development of medium-molecular-weight drugs (peptide- or oligonucleotide-based drugs) and high-molecular-weight drugs (antibody-based drugs) provide alternative pathways to preventing the formation of abnormal tau. Of particular interest are recent studies suggesting that tau droplet formation by liquid-liquid phase separation may be the initial step in aberrant tau aggregation, as well results that implicate roles for tau in dendritic and nuclear functions. Here, we review the mechanisms through which drugs can target tau and consider recent clinical trials for the treatment of tauopathies. In addition, we discuss the utility of these newer strategies and propose future directions for research on tau-targeted therapeutics.
Collapse
Affiliation(s)
- Yoshiyuki Soeda
- Laboratory for Alzheimer's Disease, Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| | - Akihiko Takashima
- Laboratory for Alzheimer's Disease, Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| |
Collapse
|