1
|
Chen K, Morizawa YM, Nuriel T, Al-Dalahmah O, Xie Z, Yang G. Selective removal of astrocytic PERK protects against glymphatic impairment and decreases toxic aggregation of β-amyloid and tau. Neuron 2025:S0896-6273(25)00310-1. [PMID: 40403715 DOI: 10.1016/j.neuron.2025.04.027] [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: 05/03/2024] [Revised: 01/02/2025] [Accepted: 04/25/2025] [Indexed: 05/24/2025]
Abstract
Dysfunction of the glymphatic system, a brain-wide waste clearance network, is strongly linked to Alzheimer's disease (AD) and the accumulation of β-amyloid (Aβ) and tau proteins. Here, we identify an astrocytic signaling pathway that can be targeted to preserve glymphatic function and mitigate neurotoxic protein buildup. Analysis of astrocytes from both human AD brains and two transgenic mouse models (5XFAD and PS19) reveals robust activation of the protein kinase RNA-like endoplasmic reticulum (ER) kinase (PERK)-α subunit of eukaryotic initiation factor 2 (eIF2α) branch of the unfolded protein response. Chronic PERK activation suppresses astrocytic protein synthesis and, through casein kinase 2 (CK2)-dependent mechanisms, disrupts the perivascular localization of aquaporin-4 (AQP4), a water channel essential for glymphatic flow. Importantly, astrocyte-specific PERK deletion or pharmacological inhibition restores AQP4 localization, enhances glymphatic clearance, reduces Aβ and tau pathology, and improves cognitive performance in mice. These findings highlight the critical role of the astrocytic PERK-CK2-AQP4 axis in glymphatic dysfunction and AD pathogenesis, positioning this pathway as a promising therapeutic target.
Collapse
Affiliation(s)
- Kai Chen
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Yosuke M Morizawa
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tal Nuriel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Osama Al-Dalahmah
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | - Guang Yang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
2
|
Tucker JK, Da Silva IIN, Gage FH. Scaffold hopping and sidechain modification from a flavone scaffold lead to discovery of potent, selective CK2A2 inhibitors with favorable properties for CNS activity. Bioorg Med Chem 2025; 127:118196. [PMID: 40367913 DOI: 10.1016/j.bmc.2025.118196] [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: 02/14/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025]
Abstract
The human protein kinase CK2 has long been of interest as a target in oncology, but new evidence is emerging of its role in central nervous system (CNS) disorders. The CK2 catalytic subunit paralog CK2A2 is enriched in the CNS relative to other tissues; however, the catalytic subunit paralog CK2A1 is expressed at similar levels across most tissues. Current treatment modalities centered on CK2 inhibition under clinical study lack evidence of CNS activity, kinome-wide selectivity, or CK2A2 paralog selectivity. Therefore, a brain-penetrant inhibitor selective for CK2A2 over CK2A1 may enable the further elucidation of the role of this paralog in CNS pathologies. In this work, we describe a series of flavone-inspired inhibitors of CK2A1 and CK2A2 designed and synthesized in a structure-based drug-design campaign. Multiple candidates demonstrating promising CK2A2 potency, kinome-wide selectivity, and CNS permeability/activity were identified, with compound 65 representing the best confluence of these properties. Although these lead candidates exhibited only up to three-fold preferential inhibition of CK2A2, they represent a starting point for the study of CK2A2 and the disorders it may mediate in the CNS.
Collapse
Affiliation(s)
- James K Tucker
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States.
| | - Ioana I N Da Silva
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
3
|
Hadzibegovic S, Bontempi B, Nicole O. Investigating the Impact of NMDA Receptor Organization and Biological Sex in the APPswe/PS1dE9 Mouse Model of Alzheimer's Disease. Int J Mol Sci 2025; 26:1737. [PMID: 40004200 PMCID: PMC11855313 DOI: 10.3390/ijms26041737] [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: 12/18/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by memory loss and cognitive decline, with women being disproportionately affected in both prevalence and severity. A key feature of AD is synaptic loss, particularly around amyloid-β (Aβ) aggregates, which correlates strongly with the severity of dementia. Oligomeric Aβ is believed to be the primary driver of synaptic dysfunction by impairing excitatory neurotransmission through interactions with synaptic receptors, including N-methyl-D-aspartate (NMDA) receptors. However, the influence of sex on these synaptic changes and NMDA receptor mislocalization in AD is not well understood. This study examined potential sex-specific differences in synaptotoxicity and the role of extrasynaptic GluN2B-containing NMDA receptors in AD pathogenesis using the APP/PS1 double transgenic mouse model. Although both male and female mice showed a similar amyloid burden and cognitive impairments, synaptic alterations were slightly less severe in females, suggesting subtle sex differences in synaptic pathology. Both sexes exhibited the mislocalization of GluN2B subunits to extrasynaptic areas, which was linked to reduced PSD-95 levels and the synaptic accumulation of Aβ1-42. Intrahippocampal injections of DL-TBOA confirmed the role of extrasynaptic GluN2B-containing NMDA receptors in memory dysfunction. These findings emphasize the importance of targeting synaptic receptor trafficking to address AD-related memory deficits, potentially offering a therapeutic approach for both sexes.
Collapse
Affiliation(s)
- Senka Hadzibegovic
- Neurocentre Magendie, INSERM U1215, 33077 Bordeaux, France;
- University of Bordeaux, 33077 Bordeaux, France;
| | - Bruno Bontempi
- University of Bordeaux, 33077 Bordeaux, France;
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine, CNRS UMR 5287, 33000 Bordeaux, France
| | - Olivier Nicole
- University of Bordeaux, 33077 Bordeaux, France;
- Institut Interdisciplinaire de Neurosciences, CNRS, UMR 5297, 33077 Bordeaux, France
| |
Collapse
|
4
|
Fang L, Xue H, Lin Z, Pan W. Multivariate proteome-wide association study to identify causal proteins for Alzheimer disease. Am J Hum Genet 2025; 112:291-300. [PMID: 39793580 PMCID: PMC11866954 DOI: 10.1016/j.ajhg.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/13/2025] Open
Abstract
Alzheimer disease (AD) is a complex and progressive neurodegenerative disorder that accounts for the majority of individuals with dementia. Here, we aim to identify causal plasma proteins for AD, shedding light on the etiology of AD. We utilized the latest large-scale plasma proteomic data from the UK Biobank Pharma Proteomics Project (UKB-PPP) and AD genome-wide association study (GWAS) summary data from the International Genomics of Alzheimer's Project (IGAP). Via a robust univariate instrumental variable (IV) regression method, we identified causal proteins through cis-protein quantitative trait loci (pQTLs) and (both cis- and trans-)pQTLs. To further reduce potential false positives due to high linkage disequilibrium (LD) of some pQTLs and high correlations among some proteins, we developed a robust multivariate IV regression method, called two-stage constrained maximum likelihood (MV-2ScML), to distinguish direct and confounding/mediating effects of proteins; some key features of the method include its robustness to invalid IVs and applicability to GWAS summary data. Our work highlights some differences between using cis-pQTLs and trans-pQTLs and critical values of multivariate analysis for fine-mapping causal proteins, providing insights into plasma protein pathways to AD.
Collapse
Affiliation(s)
- Lei Fang
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, MN, USA
| | - Haoran Xue
- Department of Biostatistics, City University of Hong Kong, Kowloon, Hong Kong
| | - Zhaotong Lin
- Department of Statistics, Florida State University, Tallahassee, FL, USA
| | - Wei Pan
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Peng CH, Hwang TL, Hung SC, Tu HJ, Tseng YT, Lin TE, Lee CC, Tseng YC, Ko CY, Yen SC, Hsu KC, Pan SL, HuangFu WC. Identification, biological evaluation, and crystallographic analysis of coumestrol as a novel dual-specificity tyrosine-phosphorylation-regulated kinase 1A inhibitor. Int J Biol Macromol 2024; 282:136860. [PMID: 39481728 DOI: 10.1016/j.ijbiomac.2024.136860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
Alzheimer's disease (AD) is an irreversible neurodegenerative disease, with tau pathology caused by abnormally activated dual-specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A) being one of the culprits. Coumestrol, a phytoestrogen and natural antioxidant found in various plants, has been reported to alleviate AD, but the underlying mechanism remains unclear. We confirmed coumestrol as a novel DYRK1A inhibitor through enzyme-based assays, X-ray crystallography, and cell line experiments. Coumestrol exhibited minimal cytotoxicity at concentrations up to 100 μM in cell types such as N2A and SH-SY5Y and reduced DYRK1A-induced phosphorylated tau protein levels by >50 % at 60 μM. In the tau protein phosphorylation and microtubule assembly assay, coumestrol at 30 μM reduced phosphorylated tau by >50 % and restored the microtubule assembly process. Coumestrol also significantly reduced amyloid-β (Aβ)-induced oxidative stress in microglia at 1 μM. In zebrafish larvae co-overexpressing DYRK1A and tau, coumestrol mitigated neuronal damage and protected motor function at 48 h-postfertilization. Our results suggest that coumestrol has potential therapeutic effects in AD by inhibiting DYRK1A, lowering p-Tau levels, restoring microtubule assembly, and protecting microglia cells from Aβ-induced cell death, providing new insights into the development of coumestrol as a potential AD treatment.
Collapse
Affiliation(s)
- Chao-Hsiang Peng
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Tsong-Long Hwang
- Research Center for Chinese Herbal Medicine and Graduate Institute of Healthy Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shao-Chi Hung
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Animal Science and Technology, National Taiwan University, Taiwan
| | - Huang-Ju Tu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yen-Tzu Tseng
- Department of Animal Science and Technology, National Taiwan University, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Chung Lee
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chi Tseng
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chiung-Yuan Ko
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 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
|
6
|
Greco FA, Krämer A, Wahl L, Elson L, Ehret TAL, Gerninghaus J, Möckel J, Müller S, Hanke T, Knapp S. Synthesis and evaluation of chemical linchpins for highly selective CK2α targeting. Eur J Med Chem 2024; 276:116672. [PMID: 39067440 DOI: 10.1016/j.ejmech.2024.116672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024]
Abstract
Casein kinase-2 (CK2) are serine/threonine kinases with dual co-factor (ATP and GTP) specificity, that are involved in the regulation of a wide variety of cellular functions. Small molecules targeting CK2 have been described in the literature targeting different binding pockets of the kinase with a focus on type I inhibitors such as the recently published chemical probe SGC-CK2-1. In this study, we investigated whether known allosteric inhibitors binding to a pocket adjacent to helix αD could be combined with ATP mimetic moieties defining a novel class of ATP competitive compounds with a unique binding mode. Linking both binding sites requires a chemical linking moiety that would introduce a 90-degree angle between the ATP mimetic ring system and the αD targeting moiety, which was realized using a sulfonamide. The synthesized inhibitors were highly selective for CK2 with binding constants in the nM range and low micromolar activity. While these inhibitors need to be further improved, the present work provides a structure-based design strategy for highly selective CK2 inhibitors.
Collapse
Affiliation(s)
- Francesco A Greco
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), DKTK Site Frankfurt-Mainz, 69120 Heidelberg, Germany
| | - Laurenz Wahl
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Lewis Elson
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Theresa A L Ehret
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Joshua Gerninghaus
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Janina Möckel
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany
| | - Thomas Hanke
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany.
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt Am Main, Germany; Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt Am Main, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), DKTK Site Frankfurt-Mainz, 69120 Heidelberg, Germany.
| |
Collapse
|
7
|
Escamilla S, Sáez-Valero J, Cuchillo-Ibáñez I. NMDARs in Alzheimer's Disease: Between Synaptic and Extrasynaptic Membranes. Int J Mol Sci 2024; 25:10220. [PMID: 39337704 PMCID: PMC11431980 DOI: 10.3390/ijms251810220] [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: 09/02/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are glutamate receptors with key roles in synaptic communication and plasticity. The activation of synaptic NMDARs initiates plasticity and stimulates cell survival. In contrast, the activation of extrasynaptic NMDARs can promote cell death underlying a potential mechanism of neurodegeneration occurring in Alzheimer's disease (AD). The distribution of synaptic versus extrasynaptic NMDARs has emerged as an important parameter contributing to neuronal dysfunction in neurodegenerative diseases including AD. Here, we review the concept of extrasynaptic NMDARs, as this population is present in numerous neuronal cell membranes but also in the membranes of various non-neuronal cells. Previous evidence regarding the membranal distribution of synaptic versus extrasynaptic NMDRs in relation to AD mice models and in the brains of AD patients will also be reviewed.
Collapse
Affiliation(s)
- Sergio Escamilla
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550 Sant Joan d’Alacant, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Javier Sáez-Valero
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550 Sant Joan d’Alacant, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Inmaculada Cuchillo-Ibáñez
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550 Sant Joan d’Alacant, Spain;
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), 03550 Sant Joan d’Alacant, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
8
|
Soares C, Da Ros LU, Machado LS, Rocha A, Lazzarotto G, Carello-Collar G, De Bastiani MA, Ferrari-Souza JP, Lussier FZ, Souza DO, Rosa-Neto P, Pascoal TA, Bellaver B, Zimmer ER. The glutamatergic system in Alzheimer's disease: a systematic review with meta-analysis. Mol Psychiatry 2024; 29:2261-2273. [PMID: 38366114 DOI: 10.1038/s41380-024-02473-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/18/2024]
Abstract
Glutamatergic neurotransmission system dysregulation may play an important role in the pathophysiology of Alzheimer's disease (AD). However, reported results on glutamatergic components across brain regions are contradictory. Here, we conducted a systematic review with meta-analysis to examine whether there are consistent glutamatergic abnormalities in the human AD brain. We searched PubMed and Web of Science (database origin-October 2023) reports evaluating glutamate, glutamine, glutaminase, glutamine synthetase, glutamate reuptake, aspartate, excitatory amino acid transporters, vesicular glutamate transporters, glycine, D-serine, metabotropic and ionotropic glutamate receptors in the AD human brain (PROSPERO #CDRD42022299518). The studies were synthesized by outcome and brain region. We included cortical regions, the whole brain (cortical and subcortical regions combined), the entorhinal cortex and the hippocampus. Pooled effect sizes were determined with standardized mean differences (SMD), random effects adjusted by false discovery rate, and heterogeneity was examined by I2 statistics. The search retrieved 6 936 articles, 63 meeting the inclusion criteria (N = 709CN/786AD; mean age 75/79). We showed that the brain of AD individuals presents decreased glutamate (SMD = -0.82; I2 = 74.54%; P < 0.001) and aspartate levels (SMD = -0.64; I2 = 89.71%; P = 0.006), and reuptake (SMD = -0.75; I2 = 83.04%; P < 0.001. We also found reduced α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPAR)-GluA2/3 levels (SMD = -0.63; I2 = 95.55%; P = 0.046), hypofunctional N-methyl-D-aspartate receptor (NMDAR) (SMD = -0.60; I2 = 91.47%; P < 0.001) and selective reduction of NMDAR-GluN2B subunit levels (SMD = -1.07; I2 = 41.81%; P < 0.001). Regional differences include lower glutamate levels in cortical areas and aspartate levels in cortical areas and in the hippocampus, reduced glutamate reuptake, reduced AMPAR-GluA2/3 in the entorhinal cortex, hypofunction of NMDAR in cortical areas, and a decrease in NMDAR-GluN2B subunit levels in the entorhinal cortex and hippocampus. Other parameters studied were not altered. Our findings show depletion of the glutamatergic system and emphasize the importance of understanding glutamate-mediated neurotoxicity in AD. This study has implications for the development of therapies and biomarkers in AD.
Collapse
Affiliation(s)
- Carolina Soares
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lucas Uglione Da Ros
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Luiza Santos Machado
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Andreia Rocha
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Gabriela Lazzarotto
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Giovanna Carello-Collar
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Marco A De Bastiani
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Porto Alegre, Brazil
| | - João Pedro Ferrari-Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Firoza Z Lussier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC, Canada
- Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux de l'Ouest-de-l'Île-de-Montréal, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruna Bellaver
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, UFRGS, Porto Alegre, Brazil.
- Department of Biochemistry, UFRGS, Porto Alegre, Brazil.
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.
- Brain Institute of Rio Grande do Sul - Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.
- Department of Pharmacology, UFRGS, Porto Alegre, Brazil.
| |
Collapse
|
9
|
Liu Y, Xia D, Zhong L, Chen L, Zhang L, Ai M, Mei R, Pang R. Casein Kinase 2 Affects Epilepsy by Regulating Ion Channels: A Potential Mechanism. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:894-905. [PMID: 37350003 DOI: 10.2174/1871527322666230622124618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 06/24/2023]
Abstract
Epilepsy, characterized by recurrent seizures and abnormal brain discharges, is the third most common chronic disorder of the Central Nervous System (CNS). Although significant progress has been made in the research on antiepileptic drugs (AEDs), approximately one-third of patients with epilepsy are refractory to these drugs. Thus, research on the pathogenesis of epilepsy is ongoing to find more effective treatments. Many pathological mechanisms are involved in epilepsy, including neuronal apoptosis, mossy fiber sprouting, neuroinflammation, and dysfunction of neuronal ion channels, leading to abnormal neuronal excitatory networks in the brain. CK2 (Casein kinase 2), which plays a critical role in modulating neuronal excitability and synaptic transmission, has been shown to be associated with epilepsy. However, there is limited research on the mechanisms involved. Recent studies have suggested that CK2 is involved in regulating the function of neuronal ion channels by directly phosphorylating them or their binding partners. Therefore, in this review, we will summarize recent research advances regarding the potential role of CK2 regulating ion channels in epilepsy, aiming to provide more evidence for future studies.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Di Xia
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Lianmei Zhong
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Ling Chen
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
- Yunnan Provincial Clinical Research Center for Neurological Disease, Kunming, Yunnan, 650032, China
| | - Linming Zhang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Mingda Ai
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Rong Mei
- Department of Neurology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, 650034, China
| | - Ruijing Pang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| |
Collapse
|
10
|
Liu W, Li Y, Zhao T, Gong M, Wang X, Zhang Y, Xu L, Li W, Li Y, Jia J. The role of N-methyl-D-aspartate glutamate receptors in Alzheimer's disease: From pathophysiology to therapeutic approaches. Prog Neurobiol 2023; 231:102534. [PMID: 37783430 DOI: 10.1016/j.pneurobio.2023.102534] [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: 02/27/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
N-Methyl-D-aspartate glutamate receptors (NMDARs) are involved in multiple physiopathological processes, including synaptic plasticity, neuronal network activities, excitotoxic events, and cognitive impairment. Abnormalities in NMDARs can initiate a cascade of pathological events, notably in Alzheimer's disease (AD) and even other neuropsychiatric disorders. The subunit composition of NMDARs is plastic, giving rise to a diverse array of receptor subtypes. While they are primarily found in neurons, NMDAR complexes, comprising both traditional and atypical subunits, are also present in non-neuronal cells, influencing the functions of various peripheral tissues. Furthermore, protein-protein interactions within NMDAR complexes has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation, and mitochondrial dysfunction, all of which potentially served as an obligatory relay of cognitive impairment. Nonetheless, the precise mechanistic link remains to be fully elucidated. In this review, we provided an in-depth analysis of the structure and function of NMDAR, investigated their interactions with various pathogenic proteins, discussed the current landscape of NMDAR-based therapeutics, and highlighted the remaining challenges during drug development.
Collapse
Affiliation(s)
- Wenying Liu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Tan Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Min Gong
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Xuechu Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yue Zhang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Wenwen Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China.
| |
Collapse
|
11
|
Gómez J, Artigas L, Valls R, Gervas-Arruga J. An in silico approach to identify early damage biomarker candidates in metachromatic leukodystrophy. Mol Genet Metab Rep 2023; 35:100974. [PMID: 37275681 PMCID: PMC10233284 DOI: 10.1016/j.ymgmr.2023.100974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 06/07/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a rare, autosomal recessive lysosomal storage disease. Deficient activity of arylsulfatase A causes sulfatides to accumulate in cells of different tissues, including those in the central and peripheral nervous systems, leading to progressive demyelination and neurodegeneration. Although there is some association between specific arylsulfatase A alleles and disease severity, genotype-phenotype correlations are not fully understood. We aimed to identify biomarker candidates of early tissue damage in MLD using a modeling approach based on systems biology. A review of the literature was performed in an initial disease characterization step, allowing identification of pathophysiological processes involved in MLD and proteins relating to these processes. Three mathematical models were generated to simulate different stages of MLD at the molecular level: an early pro-inflammatory stage model (including only processes considered to be active in the early stages of disease), a pre-demyelination stage model (including additional processes that are active after some disease progression), and a demyelination stage model (in which all pathophysiological processes are active). The models evaluated 3457 proteins of interest, individually and by pairs through data mining techniques, applying five filters to prioritize biomarkers that could differentiate between the models. Sixteen potential biomarkers were identified, including effectors relating to mitochondrial dysfunction, remyelination, and neurodegeneration. The findings were corroborated in a gene expression data set from T lymphocytes of patients with MLD; all candidates formed combinations that were able to distinguish patients with MLD from controls, and all but one candidate distinguished late-infantile MLD from juvenile MLD as part of a combinatorial biomarker pair. In particular, pro-neuregulin-1 appeared as differential on all comparisons (patients with MLD vs controls and within clinical subtypes); casein kinase II subunit alpha was detected as a potential individual marker within clinical subtypes. These findings provide a panel of biomarker candidates suitable for experimental validation and highlight the utility of mathematical models to identify biomarker candidates of early tissue damage in MLD with a high degree of accuracy and sensitivity.
Collapse
|
12
|
Coliță CI, Olaru DG, Coliță D, Hermann DM, Coliță E, Glavan D, Popa-Wagner A. Induced Coma, Death, and Organ Transplantation: A Physiologic, Genetic, and Theological Perspective. Int J Mol Sci 2023; 24:ijms24065744. [PMID: 36982814 PMCID: PMC10059721 DOI: 10.3390/ijms24065744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
In the clinic, the death certificate is issued if brain electrical activity is no longer detectable. However, recent research has shown that in model organisms and humans, gene activity continues for at least 96 h postmortem. The discovery that many genes are still working up to 48 h after death questions our definition of death and has implications for organ transplants and forensics. If genes can be active up to 48 h after death, is the person technically still alive at that point? We discovered a very interesting parallel between genes that were upregulated in the brain after death and genes upregulated in the brains that were subjected to medically-induced coma, including transcripts involved in neurotransmission, proteasomal degradation, apoptosis, inflammation, and most interestingly, cancer. Since these genes are involved in cellular proliferation, their activation after death could represent the cellular reaction to escape mortality and raises the question of organ viability and genetics used for transplantation after death. One factor limiting the organ availability for transplantation is religious belief. However, more recently, organ donation for the benefit of humans in need has been seen as “posthumous giving of organs and tissues can be a manifestation of love spreading also to the other side of death”.
Collapse
Affiliation(s)
- Cezar-Ivan Coliță
- Doctoral School, University of Medicine and Pharmacy Carol Davila, 020276 Bucharest, Romania; (C.-I.C.)
| | - Denissa-Greta Olaru
- Department of Psychiatry, University for Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
| | - Daniela Coliță
- Doctoral School, University of Medicine and Pharmacy Carol Davila, 020276 Bucharest, Romania; (C.-I.C.)
| | - Dirk M. Hermann
- Chair of Vascular Neurology, Dementia and Ageing, Department of Neurology, University Hospital Essen, 45147 Essen, Germany
| | - Eugen Coliță
- Doctoral School, University of Medicine and Pharmacy Carol Davila, 020276 Bucharest, Romania; (C.-I.C.)
| | - Daniela Glavan
- Department of Psychiatry, University for Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
- Correspondence: (D.G.); (A.P.-W.)
| | - Aurel Popa-Wagner
- Department of Psychiatry, University for Medicine and Pharmacy Craiova, 200349 Craiova, Romania;
- Chair of Vascular Neurology, Dementia and Ageing, Department of Neurology, University Hospital Essen, 45147 Essen, Germany
- Correspondence: (D.G.); (A.P.-W.)
| |
Collapse
|
13
|
Mitroshina EV, Pakhomov AM, Krivonosov MI, Yarkov RS, Gavrish MS, Shkirin AV, Ivanchenko MV, Vedunova MV. Novel Algorithm of Network Calcium Dynamics Analysis for Studying the Role of Astrocytes in Neuronal Activity in Alzheimer's Disease Models. Int J Mol Sci 2022; 23:ijms232415928. [PMID: 36555569 PMCID: PMC9781291 DOI: 10.3390/ijms232415928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Accumulated experimental data strongly suggest that astrocytes play an important role in the pathogenesis of neurodegeneration, including Alzheimer's disease (AD). The effect of astrocytes on the calcium activity of neuron-astroglia networks in AD modelling was the object of the present study. We have expanded and improved our approach's capabilities to analyze calcium activity. We have developed a novel algorithm to construct dynamic directed graphs of both astrocytic and neuronal networks. The proposed algorithm allows us not only to identify functional relationships between cells and determine the presence of network activity, but also to characterize the spread of the calcium signal from cell to cell. Our study showed that Alzheimer's astrocytes can change the functional pattern of the calcium activity of healthy nerve cells. When healthy nerve cells were cocultivated with astrocytes treated with Aβ42, activation of calcium signaling was found. When healthy nerve cells were cocultivated with 5xFAD astrocytes, inhibition of calcium signaling was observed. In this regard, it seems relevant to further study astrocytic-neuronal interactions as an important factor in the regulation of the functional activity of brain cells during neurodegenerative processes. The approach to the analysis of streaming imaging data developed by the authors is a promising tool for studying the collective calcium dynamics of nerve cells.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
- Correspondence: ; Tel.: +7-950-604-5137
| | - Alexander M. Pakhomov
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
- Institute of Applied Physics RAS, 46 Ulyanov Street, Nizhny Novgorod 603950, Russia
| | - Mikhail I. Krivonosov
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
- Department of Applied Mathematics, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Roman S. Yarkov
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria S. Gavrish
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Alexey V. Shkirin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilova St. 38, Moscow 119991, Russia
- Laser Physics Department, National Research Nuclear University MEPhI, Kashirskoe Sh. 31, Moscow 115409, Russia
| | - Mikhail V. Ivanchenko
- Department of Applied Mathematics, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Maria V. Vedunova
- Department of Neurotechnology, Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| |
Collapse
|
14
|
Di Mambro A, Esposito M. Thirty years of SET/TAF1β/I2PP2A: from the identification of the biological functions to its implications in cancer and Alzheimer's disease. Biosci Rep 2022; 42:BSR20221280. [PMID: 36345878 PMCID: PMC9679398 DOI: 10.1042/bsr20221280] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/17/2022] [Accepted: 11/07/2022] [Indexed: 10/29/2023] Open
Abstract
The gene encoding for the protein SE translocation (SET) was identified for the first time 30 years ago as part of a chromosomal translocation in a patient affected by leukemia. Since then, accumulating evidence have linked overexpression of SET, aberrant SET splicing, and cellular localization to cancer progression and development of neurodegenerative tauopathies such as Alzheimer's disease. Molecular biology tools, such as targeted genetic deletion, and pharmacological approaches based on SET antagonist peptides, have contributed to unveil the molecular functions of SET and its implications in human pathogenesis. In this review, we provide an overview of the functions of SET as inhibitor of histone and non-histone protein acetylation and as a potent endogenous inhibitor of serine-threonine phosphatase PP2A. We discuss the role of SET in multiple cellular processes, including chromatin remodelling and gene transcription, DNA repair, oxidative stress, cell cycle, apoptosis cell migration and differentiation. We review the molecular mechanisms linking SET dysregulation to tumorigenesis and discuss how SET commits neurons to progressive cell death in Alzheimer's disease, highlighting the rationale of exploiting SET as a therapeutic target for cancer and neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Antonella Di Mambro
- The Centre for Integrated Research in Life and Health Sciences, School of Health and Life Science, University of Roehampton, London, U.K
| | - Maria Teresa Esposito
- The Centre for Integrated Research in Life and Health Sciences, School of Health and Life Science, University of Roehampton, London, U.K
| |
Collapse
|
15
|
White A, McGlone A, Gomez-Pastor R. Protein Kinase CK2 and Its Potential Role as a Therapeutic Target in Huntington's Disease. Biomedicines 2022; 10:1979. [PMID: 36009526 PMCID: PMC9406209 DOI: 10.3390/biomedicines10081979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Huntington's Disease (HD) is a devastating neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the HTT gene, for which no disease modifying therapies are currently available. Much of the recent research has focused on developing therapies to directly lower HTT expression, and while promising, these therapies have presented several challenges regarding administration and efficacy. Another promising therapeutic approach is the modulation of HTT post-translational modifications (PTMs) that are dysregulated in disease and have shown to play a key role in HTT toxicity. Among all PTMs, modulation of HTT phosphorylation has been proposed as an attractive therapeutic option due to the possibility of orally administering specific kinase effectors. One of the kinases described to participate in HTT phosphorylation is Protein Kinase CK2. CK2 has recently emerged as a target for the treatment of several neurological and psychiatric disorders, although its role in HD remains controversial. While pharmacological studies in vitro inhibiting CK2 resulted in reduced HTT phosphorylation and increased toxicity, genetic approaches in mouse models of HD have provided beneficial effects. In this review we discuss potential therapeutic approaches related to the manipulation of HTT-PTMs with special emphasis on the role of CK2 as a therapeutic target in HD.
Collapse
Affiliation(s)
| | | | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|