1
|
Wang T, Wang B, Lu Z, Li T. Low RYR2 Level Relates to Poor Prognosis of Patients With Lung Adenocarcinoma by Promoting Tumor Cell Proliferation and Inhibiting Immune Cell Infiltration. Biotechnol Appl Biochem 2025:e2759. [PMID: 40200715 DOI: 10.1002/bab.2759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/23/2025] [Indexed: 04/10/2025]
Abstract
Ryanodine receptor type 2 (RYR2) is a large calcium channel that has been identified as one of the most frequently mutated genes in lung adenocarcinoma (LUAD). Despite its potential significance, the role of RYR2 in LUAD remains poorly understood. In this study, we obtained transcriptomic data (normal n = 59, tumor n = 541) from TCGA portal and RYR2 protein abundance data from cProSite, which includes 86 normal and 91 tumor samples. Additionally, we assembled a cohort of 38 patients with LUAD and collected paired tumor and adjacent non-tumor control samples. To investigate the functional impact of RYR2, we employed 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry analysis to assess cell viability and apoptosis, respectively. While mitochondria function was evaluated via measuring oxygen consumption rate. The relationship between RYR2 expression level and immune cell infiltration was analyzed by immunohistochemistry and flow cytometry analysis. Furthermore, RT-qPCR and enzyme-linked immunosorbent assay were used to quantify the expression levels of CCL14 and CXCL12. Our findings demonstrated that both the mRNA and protein levels of RYR2 were significantly downregulated in LUAD samples, and lower RYR2 levels are associated with the poor patient prognosis. Overexpression of RYR2 in A549 and H1299 cells resulted in impaired mitochondrial function, decreased cell viability, and increased apoptosis. Notably, RYR2 levels exhibited a negative correlation with tumor purity, and tumors with lower RYR2 levels showed diminished infiltration of T cells and dendritic cells. Knockdown of RYR2 in LUAD cells inhibited the production of chemokines, particularly CCL14 and CXCL12. In conclusion, our study reveals that RYR2 functions as a tumor suppressor in LUAD by inducing mitochondrial dysfunction and promoting immune cell infiltration.
Collapse
Affiliation(s)
- Tao Wang
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Baozhen Wang
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zhongting Lu
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Tao Li
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
2
|
Gleitze S, Ramírez OA, Vega-Vásquez I, Stefan E, Bengtson CP, Paula-Lima A, Bading H, Hidalgo C. Calcium release via IP 3R/RyR channels contributes to the nuclear and mitochondrial Ca 2+ signals elicited by neuronal stimulation. Biochem Biophys Res Commun 2025; 754:151445. [PMID: 40022811 DOI: 10.1016/j.bbrc.2025.151445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
The brain constantly adapts to environmental changes by modifying the expression of genes that enable synaptic plasticity, learning and memory. The expression of several of these genes requires nuclear calcium (Ca2+) signals, which in turn requires that Ca2+ signals generated by neuronal activity at the synapses or the soma propagate to the nucleus. Since cytoplasmic Ca2+ diffusion is highly restricted, Ca2+ signal propagation to the nucleus requires the participation of other cellular mechanisms. The inositol trisphosphate receptor (IP3R) and the ryanodine receptor (RyR) channels, both of which reside in the endoplasmic reticulum (ER) membrane, play key roles in cellular Ca2+ signal generation. Yet, their roles in the generation of nuclear and mitochondrial Ca2+ signals induced by neuronal activity require further investigation. Here, the impact of IP3R1 or RyR2 knockdown on gabazine-induced nuclear and mitochondrial Ca2+ signals in neurons was evaluated. To this aim, recombinant adeno-associated viruses (rAAVs) were used to introduce small hairpin RNAs (shRNAs) to knockdown type-1 (IP3R1) and type-2 (RyR2) channel expression in cultured rat hippocampal neurons. Additionally, synaptic contact numbers were assessed through immunocytochemistry. Knockdown of IP3R1 or RyR2 channels significantly reduced their protein contents and the generation of gabazine-induced nuclear and mitochondrial Ca2+ signals, without altering synaptic contact numbers. Our results highlight the contribution of IP3R1 and RyR2 channels to the generation of nuclear and mitochondrial Ca2+ signal induced by neuronal activity, reinforcing the role that these Ca2+ release channels play in hippocampal synaptic plasticity and memory formation.
Collapse
Affiliation(s)
- Silvia Gleitze
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, 8380453, Chile
| | - Omar A Ramírez
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
| | - Ignacio Vega-Vásquez
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, 8380453, Chile; Advanced Scientific Equipment Network (REDECA), Faculty of Medicine, Universidad de Chile, Santiago, 8380453, Chile; Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Emely Stefan
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
| | - Andrea Paula-Lima
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, 8380453, Chile; Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, 8380453, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, 8380453, Chile
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120, Heidelberg, Germany
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, 8380453, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, 8380453, Chile; Physiology and Biophysics Program, Institute of Biomedical Sciences and Center for Exercise, Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago, 8380453, Chile.
| |
Collapse
|
3
|
Zhang X, Zhang Y, Zhang T, Wang J, Liu C, Shang Q, Wei X, Zhu H, Shen H, Sun B. HCN2 deficiency correlates with memory deficits and hyperexcitability of dCA1 pyramidal neurons in Alzheimer's disease. Alzheimers Res Ther 2025; 17:55. [PMID: 40016780 PMCID: PMC11866685 DOI: 10.1186/s13195-025-01704-y] [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: 09/18/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Abnormal excitability of hippocampal neurons may lead to dysfunction of neural circuits and then causes cognitive impairments in Alzheimer's disease (AD). However, the underlying mechanisms remain to be fully elucidated. METHODS Electrophysiology was performed to examine the intrinsic excitability of CA1 neurons and the activity of the hyperpolarization-activated cyclic nucleotide-gated ion channels (HCNs) of CA1 neurons in wild type (WT) and hAPP-J20 mice. The activity of CA1 pyramidal neurons (PNs) was modulated with chemogenetics. The activity of HCNs was regulated with nonselective facilitator (cAMP) or inhibitor (ZD7288) of HCNs. Immunohistochemical staining or western blotting were performed to examine the expression of HCN1 and HCN2 in the hippocampus of WT and hAPP-J20 mice, or AD patients and non-AD controls. AAVs were injected to specifically modulate the expression of HCN2 in dorsal CA1 (dCA1) PNs. Cognitive performance of mice was assessed with behavioral tests. RESULTS dCA1 PNs were more excitable in hAPP-J20 mice, but the excitability of PNs in the ventral CA1 (vCA1) or PV neurons was comparable between WT and hAPP-J20 mice. The activity of the HCNs was reduced in dCA1 PNs of hAPP-J20 mice, and pharmacologically increasing the activity of HCNs attenuated the hyperexcitability of dCA1 PNs in hAPP-J20 mice, suggesting that the reduced activity of HCNs is associated with the hyperexcitability of dCA1 PNs in hAPP-J20 mice. The expression of HCN2 but not HCN1 was reduced in the hippocampus of hAPP-J20 mice, and the expression of HCN2 was also reduced in the hippocampus of AD patients, suggesting that dysregulation of HCN2 is associated with the reduced activity of HCNs in AD. Overexpressing HCN2 rescued the activity of HCNs, attenuated the hyperexcitability of dCA1 PNs and improved memory of hAPP-J20 mice, and knocking down HCN2 impaired the function of HCNs, increased the excitability of dCA1 PNs and led to memory deficits in WT mice. CONCLUSIONS Our data suggest that dysregulation of HCNs, particularly HCN2, contributes to the abnormal excitability of CA1 PNs in AD mice and probably in AD patients as well, and thus provide new insights into the mechanisms underlying the aberrant activity or excitability of hippocampal neurons in AD.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China.
| | - Yiping Zhang
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Ting Zhang
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Jing Wang
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Chang Liu
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China
| | - Qing Shang
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 59 Liuting Street, Haishu District, Ningbo, Zhejiang Province, 315211, China
| | - Xiaojie Wei
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China
| | - Huaqiang Zhu
- Zhejiang Pharmaceutical College, Ningbo, Zhejiang Province, 315100, China
| | - Haowei Shen
- Department of Pharmacology, Health Science Center of Ningbo University, Ningbo, Zhejiang Province, 315211, China.
| | - Binggui Sun
- Department of Anesthesiology of the Children's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine and National Clinical Research Center for Child Health; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310058, China.
| |
Collapse
|
4
|
Jin X, He W, Liu M, Wang L, Zhang Y, Xu Y, Ma L, Huang Y, Xie M. Mining functional gene modules by multi-view NMF of phenome-genome association. BMC Genomics 2025; 23:868. [PMID: 39789452 PMCID: PMC11720361 DOI: 10.1186/s12864-024-11120-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 12/02/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Mining functional gene modules from genomic data is an important step to detect gene members of pathways or other relations such as protein-protein interactions. This work explores the plausibility of detecting functional gene modules by factorizing gene-phenotype association matrix from the phenotype ontology data rather than the conventionally used gene expression data. Recently, the hierarchical structure of phenotype ontologies has not been sufficiently utilized in gene clustering while functionally related genes are consistently associated with phenotypes on the same path in phenotype ontologies. RESULTS This work demonstrates a hierarchical Nonnegative Matrix Factorization (NMF) framework, called Consistent Multi-view Nonnegative Matrix Factorization (CMNMF), which factorizes genome-phenome association matrix at consecutive levels of the hierarchical structure in phenotype ontology to mine functional gene modules. CMNMF constrains the gene clusters from the association matrices at two consecutive levels to be consistent since the genes are annotated with both the child-level phenotypes and the parent-level phenotypes in two levels. CMNMF also restricts the identified gene clusters to be densely connected in the phenotype ontology hierarchy. In the experiments on mining functionally related genes from mouse phenotype ontology and human phenotype ontology, CMNMF effectively improves clustering performance over the baseline methods. Gene ontology enrichment analysis is also conducted to verify its practical effectiveness to reveal meaningful gene modules. CONCLUSIONS Utilizing the information in the hierarchical structure of phenotype ontology, CMNMF can identify functional gene modules with more biological significance than conventional methods. CMNMF can also be a better tool for predicting members of gene pathways and protein-protein interactions.
Collapse
Affiliation(s)
- Xu Jin
- College of Software, Nankai University, TianJin, China
| | - WenQian He
- College of Software, Nankai University, TianJin, China
| | - MingMing Liu
- College of Software, Nankai University, TianJin, China
| | - Lin Wang
- College of Software, Nankai University, TianJin, China
| | - YaoGong Zhang
- College of Software, Nankai University, TianJin, China
| | - YingJie Xu
- College of Software, Nankai University, TianJin, China
| | - Ling Ma
- College of Software, Nankai University, TianJin, China
| | - YaLou Huang
- TianJin International Joint Academy of Biomedicine, TianJin, China
| | - MaoQiang Xie
- College of Software, Nankai University, TianJin, China.
| |
Collapse
|
5
|
Li J, Liu Y, Yin C, Zeng Y, Mei Y. Structural and functional remodeling of neural networks in β-amyloid driven hippocampal hyperactivity. Ageing Res Rev 2024; 101:102468. [PMID: 39218080 DOI: 10.1016/j.arr.2024.102468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Early detection of Alzheimer's disease (AD) is essential for improving the patients outcomes and advancing our understanding of disease, allowing for timely intervention and treatment. However, accurate biomarkers are still lacking. Recent evidence indicates that hippocampal hyperexcitability precedes the diagnosis of AD decades ago, can predict cognitive decline. Thus, could hippocampal hyperactivity be a robust biomarker for early-AD, and what drives hippocampal hyperactivity in early-AD? these critical questions remain to be answered. Increasing clinical and experimental studies suggest that early hippocampal activation is closely associated with longitudinal β-amyloid (Aβ) accumulation, Aβ aggregates, in turn, enhances hippocampal activity. Therefore, in this narrative review, we discuss the role of Aβ-induced altered intrinsic neuronal properties as well as structural and functional remodeling of glutamatergic, GABAergic, cholinergic, noradrenergic, serotonergic circuits in hippocampal hyperactivity. In addition, we analyze the available therapies and trials that can potentially be used clinically to attenuate hippocampal hyperexcitability in AD. Overall, the present review sheds lights on the mechanism behind Aβ-induced hippocampal hyperactivity, and highlights that hippocampal hyperactivity could be a robust biomarker and therapeutic target in prodromal AD.
Collapse
Affiliation(s)
- Jinquan Li
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yanjun Liu
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Chuhui Yin
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yan Zeng
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
6
|
Yin Q, Aryal SP, Song Y, Fu X, Richards CI. Quantitative Single-Molecule Analysis of Ryanodine Receptor 2 Subunit Assembly in Cardiac and Neuronal Tissues. Anal Chem 2024; 96:16298-16306. [PMID: 39359032 DOI: 10.1021/acs.analchem.4c03314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
We developed a method for ex vivo receptor encapsulation and single-molecule imaging techniques from neuronal and cardiac tissues, illustrating the method's broad applicability for measuring membrane receptor assembly. Ryanodine receptor 2 (RyR2) is a tetrameric Ca2+ channel governing intracellular Ca2+ dynamics, which is critical for muscle contraction. Employing GFP-RyR2 knock-in mice, we isolated individual receptor proteins in tissue-specific nanovesicles and performed subunit counting analyses to yield quantitative assessment of stoichiometric distributions across different organs. With this method, we explored the potential heterogeneity of brain-derived RyR2, which has been reported to form heteromeric assemblies with other ryanodine receptor isoforms.
Collapse
Affiliation(s)
- Qianye Yin
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Surya P Aryal
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Yongwook Song
- Computational Sciences, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Xu Fu
- Light Microscopy Center, University of Kentucky, Lexington, Kentucky 40508, United States
| | - Christopher I Richards
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| |
Collapse
|
7
|
Hori A, Inaba H, Hato T, Tanaka K, Sato S, Okamoto M, Horiuchi Y, Paran FJ, Tabe Y, Mori S, Rosales C, Akamatsu W, Murayama T, Kurebayashi N, Sakurai T, Ai T, Miida T. Carvedilol suppresses ryanodine receptor-dependent Ca2+ bursts in human neurons bearing PSEN1 variants found in early onset Alzheimer's disease. PLoS One 2024; 19:e0291887. [PMID: 39173065 PMCID: PMC11341060 DOI: 10.1371/journal.pone.0291887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 05/02/2024] [Indexed: 08/24/2024] Open
Abstract
Seizures are increasingly being recognized as the hallmark of Alzheimer's disease (AD). Neuronal hyperactivity can be a consequence of neuronal damage caused by abnormal amyloid β (Aß) depositions. However, it can also be a cell-autonomous phenomenon causing AD by Aß-independent mechanisms. Various studies using animal models have shown that Ca2+ is released from the endoplasmic reticulum (ER) via type 1 inositol triphosphate receptors (InsP3R1s) and ryanodine receptors (RyRs). To investigate which is the main pathophysiological mechanism in human neurons, we measured Ca2+ signaling in neural cells derived from three early-onset AD patients harboring Presenilin-1 variants (PSEN1 p.A246E, p.L286V, and p.M146L). Of these, it has been reported that PSEN1 p.A246E and p.L286V did not produce a significant amount of abnormal Aß. We found all PSEN1-mutant neurons, but not wild-type, caused abnormal Ca2+-bursts in a manner dependent on the calcium channel, Ryanodine Receptor 2 (RyR2). Indeed, carvedilol, an RyR2 inhibitor, and VK-II-86, an analog of carvedilol without the β-blocking effects, sufficiently eliminated the abnormal Ca2+ bursts. In contrast, Dantrolene, an inhibitor of RyR1 and RyR3, and Xestospongin c, an IP3R inhibitor, did not attenuate the Ca2+-bursts. The Western blotting showed that RyR2 expression was not affected by PSEN1 p.A246E, suggesting that the variant may activate the RyR2. The RNA-Seq data revealed that ER-stress responsive genes were increased, and mitochondrial Ca2+-transporter genes were decreased in PSEN1A246E cells compared to the WT neurons. Thus, we propose that aberrant Ca2+ signaling is a key link between human pathogenic PSEN1 variants and cell-intrinsic hyperactivity prior to deposition of abnormal Aß, offering prospects for the development of targeted prevention strategies for at-risk individuals.
Collapse
Affiliation(s)
- Atsushi Hori
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, Chiba, Japan
| | - Haruka Inaba
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takashi Hato
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Kimie Tanaka
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shoichi Sato
- Department of Clinical Engineering, Faculty of Medical Science, Juntendo University, Chiba, Japan
| | - Mizuho Okamoto
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Center for Genomic and Regenerative Medicine, School of Medicine, Juntendo University, Tokyo, Japan
| | - Yuna Horiuchi
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, Chiba, Japan
| | - Faith Jessica Paran
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shusuke Mori
- Department of Acute Care and Disaster Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Corina Rosales
- Center for Bioenergetics and the Department of Medicine, Houston Methodist Research Institute, Texas, United States of America
- Weill Cornell Medicine, New York, New York, United States of America
| | - Wado Akamatsu
- Center for Genomic and Regenerative Medicine, School of Medicine, Juntendo University, Tokyo, Japan
| | - Takashi Murayama
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, Japan
| | - Nagomi Kurebayashi
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takashi Sakurai
- Department of Pharmacology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tomohiko Ai
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Acute Care and Disaster Medicine, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Clinical Laboratory Medicine, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Takashi Miida
- Department of Clinical Laboratory Technology, Faculty of Medical Science, Juntendo University, Chiba, Japan
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Adeoye T, Shah SI, Ullah G. Systematic Analysis of Biological Processes Reveals Gene Co-expression Modules Driving Pathway Dysregulation in Alzheimer's Disease. Aging Dis 2024:AD.2024.0429. [PMID: 38913039 DOI: 10.14336/ad.2024.0429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024] Open
Abstract
Alzheimer's disease (AD) manifests as a complex systems pathology with intricate interplay among various genes and biological processes. Traditional differential gene expression (DEG) analysis, while commonly employed to characterize AD-driven perturbations, does not sufficiently capture the full spectrum of underlying biological processes. Utilizing single-nucleus RNA-sequencing data from postmortem brain samples across key regions-middle temporal gyrus, superior frontal gyrus, and entorhinal cortex-we provide a comprehensive systematic analysis of disrupted processes in AD. We go beyond the DEG-centric analysis by integrating pathway activity analysis with weighted gene co-expression patterns to comprehensively map gene interconnectivity, identifying region- and cell-type-specific drivers of biological processes associated with AD. Our analysis reveals profound modular heterogeneity in neurons and glia as well as extensive AD-related functional disruptions. Co-expression networks highlighted the extended involvement of astrocytes and microglia in biological processes beyond neuroinflammation, such as calcium homeostasis, glutamate regulation, lipid metabolism, vesicle-mediated transport, and TOR signaling. We find limited representation of DEGs within dysregulated pathways across neurons and glial cells, suggesting that differential gene expression alone may not adequately represent the disease complexity. Further dissection of inferred gene modules revealed distinct dynamics of hub DEGs in neurons versus glia, suggesting that DEGs exert more impact on neurons compared to glial cells in driving modular dysregulations underlying perturbed biological processes. Interestingly, we observe an overall downregulation of astrocyte and microglia modules across all brain regions in AD, indicating a prevailing trend of functional repression in glial cells across these regions. Notable genes from the CALM and HSP90 families emerged as hub genes across neuronal modules in all brain regions, suggesting conserved roles as drivers of synaptic dysfunction in AD. Our findings demonstrate the importance of an integrated, systems-oriented approach combining pathway and network analysis to comprehensively understand the cell-type-specific roles of genes in AD-related biological processes.
Collapse
|
9
|
Berkowitz BA, Paruchuri A, Stanek J, Abdul-Nabi M, Podolsky RH, Bustos AH, Childers KL, Murphy GG, Stangis K, Roberts R. Biomarker evidence of early vision and rod energy-linked pathophysiology benefits from very low dose DMSO in 5xFAD mice. Acta Neuropathol Commun 2024; 12:85. [PMID: 38822433 PMCID: PMC11140992 DOI: 10.1186/s40478-024-01799-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
Here, we test whether early visual and OCT rod energy-linked biomarkers indicating pathophysiology in nicotinamide nucleotide transhydrogenase (Nnt)-null 5xFAD mice also occur in Nnt-intact 5xFAD mice and whether these biomarkers can be pharmacologically treated. Four-month-old wild-type or 5xFAD C57BL/6 substrains with either a null (B6J) Nnt or intact Nnt gene (B6NTac) and 5xFAD B6J mice treated for one month with either R-carvedilol + vehicle or only vehicle (0.01% DMSO) were studied. The contrast sensitivity (CS), external limiting membrane-retinal pigment epithelium (ELM-RPE) thickness (a proxy for low pH-triggered water removal), profile shape of the hyperreflective band just posterior to the ELM (i.e., the mitochondrial configuration within photoreceptors per aspect ratio [MCP/AR]), and retinal laminar thickness were measured. Both wild-type substrains showed similar visual performance indices and dark-evoked ELM-RPE contraction. The lack of a light-dark change in B6NTac MCP/AR, unlike in B6J mice, is consistent with relatively greater mitochondrial efficiency. 5xFAD B6J mice, but not 5xFAD B6NTac mice, showed lower-than-WT CS. Light-adapted 5xFAD substrains both showed abnormal ELM-RPE contraction and greater-than-WT MCP/AR contraction. The inner retina and superior outer retina were thinner. Treating 5xFAD B6J mice with R-carvedilol + DMSO or DMSO alone corrected CS and ELM-RPE contraction but not supernormal MCP/AR contraction or laminar thinning. These results provide biomarker evidence for prodromal photoreceptor mitochondrial dysfunction/oxidative stress/oxidative damage, which is unrelated to visual performance, as well as the presence of the Nnt gene. This pathophysiology is druggable in 5xFAD mice.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA.
| | - Anuhya Paruchuri
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Josh Stanek
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Mura Abdul-Nabi
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| | - Robert H Podolsky
- Biostatistics and Study Methodology, Children's National Hospital, Silver Spring, MD, USA
| | | | | | - Geoffrey G Murphy
- Department of Molecular and Integrative Physiology, Molecular Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Katherine Stangis
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robin Roberts
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI, 48201, USA
| |
Collapse
|
10
|
Yao J, Chen SRW. RyR2-dependent modulation of neuronal hyperactivity: A potential therapeutic target for treating Alzheimer's disease. J Physiol 2024; 602:1509-1518. [PMID: 36866974 DOI: 10.1113/jp283824] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/27/2023] [Indexed: 03/04/2023] Open
Abstract
Increasing evidence suggests that simply reducing β-amyloid (Aβ) plaques may not significantly affect the progression of Alzheimer's disease (AD). There is also increasing evidence indicating that AD progression is driven by a vicious cycle of soluble Aβ-induced neuronal hyperactivity. In support of this, it has recently been shown that genetically and pharmacologically limiting ryanodine receptor 2 (RyR2) open time prevents neuronal hyperactivity, memory impairment, dendritic spine loss and neuronal cell death in AD mouse models. By contrast, increased RyR2 open probability (Po) exacerbates the onset of familial AD-associated neuronal dysfunction and induces AD-like defects in the absence of AD-causing gene mutations. Thus, RyR2-dependent modulation of neuronal hyperactivity represents a promising new target for combating AD.
Collapse
Affiliation(s)
- Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
11
|
Adeoye T, Shah SI, Ullah G. Systematic Analysis of Biological Processes Reveals Gene Co-expression Modules Driving Pathway Dysregulation in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585267. [PMID: 38559218 PMCID: PMC10980062 DOI: 10.1101/2024.03.15.585267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease (AD) manifests as a complex systems pathology with intricate interplay among various genes and biological processes. Traditional differential gene expression (DEG) analysis, while commonly employed to characterize AD-driven perturbations, does not sufficiently capture the full spectrum of underlying biological processes. Utilizing single-nucleus RNA-sequencing data from postmortem brain samples across key regions-middle temporal gyrus, superior frontal gyrus, and entorhinal cortex-we provide a comprehensive systematic analysis of disrupted processes in AD. We go beyond the DEG-centric analysis by integrating pathway activity analysis with weighted gene co-expression patterns to comprehensively map gene interconnectivity, identifying region- and cell-type-specific drivers of biological processes associated with AD. Our analysis reveals profound modular heterogeneity in neurons and glia as well as extensive AD-related functional disruptions. Co-expression networks highlighted the extended involvement of astrocytes and microglia in biological processes beyond neuroinflammation, such as calcium homeostasis, glutamate regulation, lipid metabolism, vesicle-mediated transport, and TOR signaling. We find limited representation of DEGs within dysregulated pathways across neurons and glial cells, indicating that differential gene expression alone may not adequately represent the disease complexity. Further dissection of inferred gene modules revealed distinct dynamics of hub DEGs in neurons versus glia, highlighting the differential impact of DEGs on neurons compared to glial cells in driving modular dysregulations underlying perturbed biological processes. Interestingly, we note an overall downregulation of both astrocyte and microglia modules in AD across all brain regions, suggesting a prevailing trend of functional repression in glial cells across these regions. Notable genes, including those of the CALM and HSP90 family genes emerged as hub genes across neuronal modules in all brain regions, indicating conserved roles as drivers of synaptic dysfunction in AD. Our findings demonstrate the importance of an integrated, systems-oriented approach combining pathway and network analysis for a comprehensive understanding of the cell-type-specific roles of genes in AD-related biological processes.
Collapse
Affiliation(s)
- Temitope Adeoye
- Department of Physics, University of South Florida, Tampa, FL 33620
| | - Syed I Shah
- Department of Physics, University of South Florida, Tampa, FL 33620
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620
| |
Collapse
|
12
|
Song L, Tang Y, Law BYK. Targeting calcium signaling in Alzheimer's disease: challenges and promising therapeutic avenues. Neural Regen Res 2024; 19:501-502. [PMID: 37721273 PMCID: PMC10581553 DOI: 10.4103/1673-5374.380898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 09/19/2023] Open
Affiliation(s)
- LinLin Song
- Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao Special Administration Region, China
| | - YongPei Tang
- Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao Special Administration Region, China
| | - Betty Yuen Kwan Law
- Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao Special Administration Region, China
| |
Collapse
|
13
|
Vera R, Hong N, Jiang B, Liang G, Eckenhoff MF, Kincaid HJ, Browne V, Chellaraj V, Gisewhite D, Greenberg M, Ranjan S, Zhu G, Wei H. Effects of Intranasal Dantrolene Nanoparticles on Brain Concentration and Behavior in PS19 Tau Transgenic Mice. J Alzheimers Dis 2024; 98:549-562. [PMID: 38393915 PMCID: PMC11178503 DOI: 10.3233/jad-231337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Background Repurposing dantrolene to treat Alzheimer's disease has been shown to be effective in amyloid transgenic mouse models but has not been examined in a model of tauopathy. Objective The effects of a nanoparticle intranasal formulation, the Eagle Research Formulation of Ryanodex (ERFR), in young adult and aged wild type and PS19 tau transgenic mice was investigated. Methods The bioavailability of intranasal ERFR was measured in 2 and 9-11-month-old C57BL/6J mice. Blood and brain samples were collected 20 minutes after a single ERFR dose, and the plasma and brain concentrations were analyzed. Baseline behavior was assessed in untreated PS19 tau transgenic mice at 6 and 9 months of age. PS19 mice were treated with intranasal ERFR, with or without acrolein (to potentiate cognitive dysfunction), for 3 months, beginning at 2 months of age. Animal behavior was examined, including cognition (cued and contextual fear conditioning, y-maze), motor function (rotarod), and olfaction (buried food test). Results The dantrolene concentration in the blood and brain decreased with age, with the decrease greater in the blood resulting in a higher brain to blood concentration ratio. The behavioral assays showed no significant changes in cognition, olfaction, or motor function in the PS19 mice compared to controls after chronic treatment with intranasal ERFR, even with acrolein. Conclusions Our studies suggest the intranasal administration of ERFR has higher concentrations in the brain than the blood in aged mice and has no serious systemic side effects with chronic use in PS19 mice.
Collapse
Affiliation(s)
- Robert Vera
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas Hong
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bailin Jiang
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Anesthesiology, Peking University People's Hospital, Beijing, China
| | - Ge Liang
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maryellen F Eckenhoff
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Halle J Kincaid
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Veron Browne
- Eagle Pharmaceuticals, Inc., Woodcliff Lake, NJ, USA
| | | | | | | | - Sudhir Ranjan
- Eagle Pharmaceuticals, Inc., Woodcliff Lake, NJ, USA
| | - Gaozhong Zhu
- Eagle Pharmaceuticals, Inc., Woodcliff Lake, NJ, USA
| | - Huafeng Wei
- Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
14
|
Haji-Ghassemi O, Chen YS, Woll K, Gurrola GB, Valdivia CR, Cai W, Li S, Valdivia HH, Van Petegem F. Cryo-EM analysis of scorpion toxin binding to Ryanodine Receptors reveals subconductance that is abolished by PKA phosphorylation. SCIENCE ADVANCES 2023; 9:eadf4936. [PMID: 37224245 PMCID: PMC10208580 DOI: 10.1126/sciadv.adf4936] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/18/2023] [Indexed: 05/26/2023]
Abstract
Calcins are peptides from scorpion venom with the unique ability to cross cell membranes, gaining access to intracellular targets. Ryanodine Receptors (RyR) are intracellular ion channels that control release of Ca2+ from the endoplasmic and sarcoplasmic reticulum. Calcins target RyRs and induce long-lived subconductance states, whereby single-channel currents are decreased. We used cryo-electron microscopy to reveal the binding and structural effects of imperacalcin, showing that it opens the channel pore and causes large asymmetry throughout the cytosolic assembly of the tetrameric RyR. This also creates multiple extended ion conduction pathways beyond the transmembrane region, resulting in subconductance. Phosphorylation of imperacalcin by protein kinase A prevents its binding to RyR through direct steric hindrance, showing how posttranslational modifications made by the host organism can determine the fate of a natural toxin. The structure provides a direct template for developing calcin analogs that result in full channel block, with potential to treat RyR-related disorders.
Collapse
Affiliation(s)
- Omid Haji-Ghassemi
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Yu Seby Chen
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Kellie Woll
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| | - Georgina B. Gurrola
- Universidad Nacional Autónoma de México, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotechnología, Cuaernavaca, Morelos 62271, Mexico
| | - Carmen R. Valdivia
- Department of Medicine and Cardiovascular Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Wenxuan Cai
- Department of Medicine and Cardiovascular Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Songhua Li
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hector H. Valdivia
- Department of Medicine and Cardiovascular Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, Life Sciences Centre, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
15
|
Zhang H, Bezprozvanny I. "Dirty Dancing" of Calcium and Autophagy in Alzheimer's Disease. Life (Basel) 2023; 13:life13051187. [PMID: 37240832 DOI: 10.3390/life13051187] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. There is a growing body of evidence that dysregulation in neuronal calcium (Ca2+) signaling plays a major role in the initiation of AD pathogenesis. In particular, it is well established that Ryanodine receptor (RyanR) expression levels are increased in AD neurons and Ca2+ release via RyanRs is augmented in AD neurons. Autophagy is important for removing unnecessary or dysfunctional components and long-lived protein aggregates, and autophagy impairment in AD neurons has been extensively reported. In this review we discuss recent results that suggest a causal link between intracellular Ca2+ signaling and lysosomal/autophagic dysregulation. These new results offer novel mechanistic insight into AD pathogenesis and may potentially lead to identification of novel therapeutic targets for treating AD and possibly other neurodegenerative disorders.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnical University, St. Petersburg 195251, Russia
| |
Collapse
|
16
|
Yao J, Ni M, Tian S, Sun B, Wang R, Paul Estillore J, Back TG, Wayne Chen SR. A Gain-of-function Mutation in the Gating Domain of ITPR1 Impairs Motor Movement and Increases Thermal and Mechanical Sensitivity. Neuroscience 2023; 522:11-22. [PMID: 37164302 DOI: 10.1016/j.neuroscience.2023.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/14/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023]
Abstract
Inositol 1,4,5-trisphosphate receptor type 1 (ITPR1) is an intracellular Ca2+ release channel important for a number of fundamental cellular functions. Consistent with its critical physiological significance, mutations in ITPR1 are associated with disease. Surprisingly, nearly all the disease-associated ITPR1 mutations characterized to date are loss of function. Despite the paucity of ITPR1 gain-of-function (GOF) mutations, enhanced ITPR1 function as a result of dysregulation by ITPR1 interacting proteins is thought to be associated with ataxia, learning and memory impairments, Alzheimer's disease (AD) progression, and chronic pain. However, direct evidence for the role of ITPR1 GOF in disease is lacking. To determine whether GOF in ITPR1 itself has pathological ramifications, we employed a newly developed mouse model expressing an ITPR1 mutation in the gating domain of the channel, D2594K, that markedly increased the channel's sensitivity to activation by IP3. Behavioral studies showed that the ITPR1-D2594K+/- mutant mice displayed motor deficits and reduced muscle strength. However, the ITPR1-D2594K+/- mutation did not significantly alter hippocampal learning and memory and did not change learning and memory impairments when crossed with the 5xFAD AD model mice. On the other hand, ITPR1-D2594K+/- mice exhibited increased sensitivity to thermal and mechanical stimulation compared to WT. Interestingly, R-carvedilol treatment attenuated the enhanced thermal and mechanical nociception in ITPR1-D2594K+/- mice. Thus, the ITPR1-D2594K+/- mutation in the channel's gating domain has a marked impact on motor movements and pain perception, but little effect on hippocampal learning and memory.
Collapse
Affiliation(s)
- Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Mingke Ni
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Shanshan Tian
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Bo Sun
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - John Paul Estillore
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Thomas G Back
- Department of Chemistry, University of Calgary, Calgary, AB, Canada, T2N 1N4
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
17
|
Hyperoside alleviates toxicity of β-amyloid via endoplasmic reticulum-mitochondrial calcium signal transduction cascade in APP/PS1 double transgenic Alzheimer's disease mice. Redox Biol 2023; 61:102637. [PMID: 36821955 PMCID: PMC9975698 DOI: 10.1016/j.redox.2023.102637] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Alzheimer's disease is a neurodegenerative disorder characterized by a decline in cognitive function. The β-amyloid (Aβ) hypothesis suggests that Aβ peptides can spontaneously aggregate into β-fragment-containing oligomers and protofibrils, and this activation of the amyloid pathway alters Ca2+ signaling in neurons, leading to neurotoxicity and thus apoptosis of neuronal cells. In our study, a blood-brain barrier crossing flavonol glycoside hyperoside was identified with anti-Aβ aggregation, BACE inhibitory, and neuroprotective effect in cellular or APP/PSEN1 double transgenic Alzheimer's disease mice model. While our pharmacokinetic data confirmed that intranasal administration of hyperoside resulted in a higher bio-availability in mice brain, further in vivo studies revealed that it improved motor deficit, spatial memory and learning ability of APP/PSEN1 mice with reducing level of Aβ plaques and GFAP in the cortex and hippocampus. Bioinformatics, computational docking and in vitro assay results suggested that hyperoside bind to Aβ and interacted with ryanodine receptors, then regulated cellular apoptosis via endoplasmic reticulum-mitochondrial calcium (Ca2+) signaling pathway. Consistently, it was confirmed that hyperoside increased Bcl2, decreased Bax and cyto-c protein levels, and ameliorated neuronal cell death in both in vitro and in vivo model. By regulating Aβ-induced cell death via regulation on Ca2+ signaling cascade and mitochondrial membrane potential, our study suggested that hyperoside may work as a potential therapeutic agent or preventive remedy for Alzheimer's disease.
Collapse
|
18
|
Chiantia G, Hidisoglu E, Marcantoni A. The Role of Ryanodine Receptors in Regulating Neuronal Activity and Its Connection to the Development of Alzheimer's Disease. Cells 2023; 12:cells12091236. [PMID: 37174636 PMCID: PMC10177020 DOI: 10.3390/cells12091236] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Research into the early impacts of Alzheimer's disease (AD) on synapse function is one of the most promising approaches to finding a treatment. In this context, we have recently demonstrated that the Abeta42 peptide, which builds up in the brain during the processing of the amyloid precursor protein (APP), targets the ryanodine receptors (RyRs) of mouse hippocampal neurons and potentiates calcium (Ca2+) release from the endoplasmic reticulum (ER). The uncontrolled increase in intracellular calcium concentration ([Ca2+]i), leading to the development of Ca2+ dysregulation events and related excitable and synaptic dysfunctions, is a consolidated hallmark of AD onset and possibly other neurodegenerative diseases. Since RyRs contribute to increasing [Ca2+]i and are thought to be a promising target for AD treatment, the goal of this review is to summarize the current level of knowledge regarding the involvement of RyRs in governing neuronal function both in physiological conditions and during the onset of AD.
Collapse
Affiliation(s)
| | - Enis Hidisoglu
- Department of Drug and Science Technology, University of Torino, Corso Raffaello 30, 10125 Torino, Italy
| | - Andrea Marcantoni
- Department of Drug and Science Technology, University of Torino, Corso Raffaello 30, 10125 Torino, Italy
- N.I.S. Center, University of Torino, 10125 Turin, Italy
| |
Collapse
|
19
|
Yang F, Zhao J, Chen G, Han H, Hu S, Wang N, Wang J, Chen Y, Zhou Z, Dai B, Hou Y, Liu Y. Design, synthesis, and evaluation of hydrazones as dual inhibitors of ryanodine receptors and acetylcholinesterases for Alzheimer's disease. Bioorg Chem 2023; 133:106432. [PMID: 36841050 DOI: 10.1016/j.bioorg.2023.106432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Abstract
Alzheimer's disease (AD) implicates neuronal loss, plaque and neurofibrillary tangle formation, and disturbed neuronal Ca2+ homeostasis, which leads to severe dementia, memory loss, as well as thinking and behavioral perturbations that could ultimately lead to death. Calcium dysregulation and low acetylcholine levels are two main mechanisms implicated in Alzheimer's disease progression. Simultaneous inhibition of calcium oscillations (store overload-induced Ca2+ release [SOICR]) and acetylcholinesterase (AChE) by a single molecule may bring a new breath of hope for AD treatment. Here, we described some dantrolene derivatives as dual inhibitors of the ryanodine receptor and AChE. Two series of acylhydrazone/sulfonylhydrazone derivatives with aromaticgroup were designed and synthesized. In this study, the target compounds were evaluated for their ability to inhibit SOICR and AChE in vitro, using dantrolene and donepezil as positive controls. Compound 22a exhibited excellent and balanced inhibitory potency against SOICR (inhibition (%) = 90.1, IC50 = 0.162 μM) and AChE (inhibition (%) = 93.5, IC50 = 0.372 μM). Docking simulations showed that several preferred compounds could bind to the active sites of both the proteins, further validating the rationality of the design strategy. Potential therapeutic effects in AD were evaluated using the Barnes maze and Morris water maze tests, which demonstrated that compound 22a significantly improved memory and cognitive behavior in AD model mice. Moreover, it was also found that compound 22a could enhance synaptic strength by measuring hippocampal long-term potentiation (LTP) in brain slices. These results suggested that the introduction of a sulfonyl-hydrazone scaffold and aromatic substitution to dantrolene derivatives provided a useful template for the development of potential chemical entities against AD.
Collapse
Affiliation(s)
- Fan Yang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Jiangang Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Guang Chen
- Department of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Hao Han
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Shuang Hu
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Ningwei Wang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Junqin Wang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Yuzhen Chen
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Zihao Zhou
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Baozhu Dai
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China
| | - Yunlei Hou
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China.
| | - Yajing Liu
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China; Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, PR China.
| |
Collapse
|
20
|
Wickline JL, Smith S, Shin R, Odfalk K, Sanchez J, Javors M, Ginsburg B, Hopp SC. L-type calcium channel antagonist isradipine age-dependently decreases plaque associated dystrophic neurites in 5XFAD mouse model. Neuropharmacology 2023; 227:109454. [PMID: 36740015 PMCID: PMC9987839 DOI: 10.1016/j.neuropharm.2023.109454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Epidemiological studies suggest that L-type calcium channel (LTCC) antagonists may reduce the incidence of age-associated neurodegenerative diseases including Alzheimer's disease (AD). However, the neuroprotective mechanism of LTCC antagonists is unknown. Amyloid-β (Aβ) pathology disrupts intracellular calcium signaling, which regulates lysosomes and microglial responses. Neurons near Aβ plaques develop dystrophic neurites, which are abnormal swellings that accumulate lysosomes. Further, microglia accumulate around Aβ plaques and secrete inflammatory cytokines. We hypothesized that antagonism of LTCCs with isradipine would reduce Aβ plaque-associated dystrophic neurites and inflammatory microglia in the 5XFAD mouse model by restoring normal intracellular calcium regulation. To test this hypothesis, we treated 6- and 9-month-old 5XFAD mice with isradipine and tested behavior, examined Aβ plaques, microglia, and dystrophic neurites. We found that isradipine treatment age-dependently reduces dystrophic neurites and leads to trending decreases in Aβ but does not modulate plaque associated microglia regardless of age. Our findings provide insight into how antagonizing LTCCs alters specific cell types in the Aβ plaque environment, providing valuable information for potential treatment targets in future AD studies.
Collapse
Affiliation(s)
- Jessica L Wickline
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Sabrina Smith
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Riley Shin
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Kristian Odfalk
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Jesse Sanchez
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Martin Javors
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Brett Ginsburg
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
21
|
Perone I, Ghena N, Wang J, Mackey C, Wan R, Malla S, Gorospe M, Cheng A, Mattson MP. Mitochondrial SIRT3 Deficiency Results in Neuronal Network Hyperexcitability, Accelerates Age-Related Aβ Pathology, and Renders Neurons Vulnerable to Aβ Toxicity. Neuromolecular Med 2023; 25:27-39. [PMID: 35749057 PMCID: PMC9810471 DOI: 10.1007/s12017-022-08713-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/01/2022] [Indexed: 01/05/2023]
Abstract
Aging is the major risk factor for Alzheimer's disease (AD). Mitochondrial dysfunction and neuronal network hyperexcitability are two age-related alterations implicated in AD pathogenesis. We found that levels of the mitochondrial protein deacetylase sirtuin-3 (SIRT3) are significantly reduced, and consequently mitochondria protein acetylation is increased in brain cells during aging. SIRT3-deficient mice exhibit robust mitochondrial protein hyperacetylation and reduced mitochondrial mass during aging. Moreover, SIRT3-deficient mice exhibit epileptiform and burst-firing electroencephalogram activity indicating neuronal network hyperexcitability. Both aging and SIRT3 deficiency result in increased sensitivity to kainic acid-induced seizures. Exposure of cultured cerebral cortical neurons to amyloid β-peptide (Aβ) results in a reduction in SIRT3 levels and SIRT3-deficient neurons exhibit heightened sensitivity to Aβ toxicity. Finally, SIRT3 haploinsufficiency in middle-aged App/Ps1 double mutant transgenic mice results in a significant increase in Aβ load compared with App/Ps1 double mutant mice with normal SIRT3 levels. Collectively, our findings suggest that SIRT3 plays an important role in protecting neurons against Aβ pathology and excitotoxicity.
Collapse
Affiliation(s)
- Isabella Perone
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Nathaniel Ghena
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Jing Wang
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
- Department of Integrative Medicine and Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chelsea Mackey
- Department of Integrative Medicine and Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Laboratory of Cardiovascular Science, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Ruiqian Wan
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Sulochan Malla
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA
| | - Aiwu Cheng
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA.
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA.
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, 21224, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
22
|
Berkowitz BA, Podolsky RH, Childers KL, Roberts R, Waseem R. Multiple Bioenergy-Linked OCT Biomarkers Suggest Greater-Than-Normal Rod Mitochondria Activity Early in Experimental Alzheimer's Disease. Invest Ophthalmol Vis Sci 2023; 64:12. [PMID: 36867132 PMCID: PMC9988708 DOI: 10.1167/iovs.64.3.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Purpose In Alzheimer's disease, central brain neurons show evidence for early hyperactivity. It is unclear if this occurs in the retina, another disease target. Here, we tested for imaging biomarker manifestation of prodromal hyperactivity in rod mitochondria in vivo in experimental Alzheimer's disease. Methods Light- and dark-adapted 4-month-old 5xFAD and wild-type (WT) mice, both on a C57BL/6J background, were studied with optical coherence tomography (OCT). We measured the reflectivity profile shape of the inner segment ellipsoid zone (EZ) as a proxy for mitochondria distribution. Two additional indices responsive to mitochondria activity were also measured: the thickness of the external limiting membrane-retinal pigment epithelium (ELM-RPE) region and the signal magnitude of a hyporeflective band (HB) between photoreceptor tips and apical RPE. Retinal laminar thickness and visual performance were evaluated. Results In response to low energy demand (light), WT mice showed the expected elongation in EZ reflectivity profile shape, relatively thicker ELM-RPE, and greater HB signal. Under high energy demand (dark), the EZ reflectivity profile shape was rounder, the ELM-RPE was thinner, and the HB was reduced. These OCT biomarker patterns for light-adapted 5xFAD mice did not match those of light-adapted WT mice but rather that of dark-adapted WT mice. Dark-adapted 5xFAD and WT mice showed the same biomarker pattern. The 5xFAD mice exhibited modest nuclear layer thinning and lower-than-normal contrast sensitivity. Conclusions Results from three OCT bioenergy biomarkers raise the novel possibility of early rod hyperactivity in vivo in a common Alzheimer's disease model.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Robert H Podolsky
- Biostatistics and Study Methodology, Children's National Hospital, Silver Spring, Maryland, United States
| | - Karen L Childers
- Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan, United States
| | - Robin Roberts
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Rida Waseem
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
23
|
Zhang H, Knight C, Chen SRW, Bezprozvanny I. A Gating Mutation in Ryanodine Receptor Type 2 Rescues Phenotypes of Alzheimer's Disease Mouse Models by Upregulating Neuronal Autophagy. J Neurosci 2023; 43:1441-1454. [PMID: 36627208 PMCID: PMC9987572 DOI: 10.1523/jneurosci.1820-22.2022] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/26/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
It is well established that ryanodine receptors (RyanRs) are overactive in Alzheimer's disease (AD), and it has been suggested that inhibition of RyanR is potentially beneficial for AD treatment. In the present study, we explored a potential connection between basal RyanR activity and autophagy in neurons. Autophagy plays an important role in clearing damaged organelles and long-lived protein aggregates, and autophagy dysregulation occurs in both AD patients and AD animal models. Autophagy is known to be regulated by intracellular calcium (Ca2+) signals, and our results indicated that basal RyanR2 activity in hippocampal neurons inhibited autophagy through activation of calcineurin and the resulting inhibition of the AMPK (AMP-activated protein kinase)-ULK1 (unc-51-like autophagy-activating kinase 1) pathway. Thus, we hypothesized that increased basal RyanR2 activity in AD may lead to the inhibition of neuronal autophagy and accumulation of β-amyloid. To test this hypothesis, we took advantage of the RyanR2-E4872Q knock-in mouse model (EQ) in which basal RyanR2 activity is reduced because of shortened channel open time. We discovered that crossing EQ mice with the APPKI and APPPS1 mouse models of AD (both males and females) rescued amyloid accumulation and LTP impairment in these mice. Our results revealed that reduced basal activity of RyanR2-EQ channels disinhibited the autophagic pathway and led to increased amyloid clearance in these models. These data indicated a potential pathogenic outcome of RyanR2 overactivation in AD and also provided additional targets for therapeutic intervention in AD. Basal activity of ryanodine receptors controls neuronal autophagy and contributes to development of the AD phenotype.SIGNIFICANCE STATEMENT It is well established that neuronal autophagy is impaired in Alzheimer's disease (AD). Our results suggest that supranormal calcium (Ca2+) release from endoplasmic reticulum contributes to the inhibition of autophagy in AD and that reduction in basal activity of type 2 ryanodine receptors disinhibits the neuronal autophagic pathway and leads to increased amyloid clearance in AD models. Our findings directly link neuronal Ca2+ dysregulation with autophagy dysfunction in AD and point to additional targets for therapeutic intervention.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas 75390
| | - Caitlynn Knight
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas 75390
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas 75390
- Laboratory of Molecular Neurodegeneration, St. Petersburg State Polytechnical Universty, St. Petersburg 195251, Russian Federation
| |
Collapse
|
24
|
Wang J, Mei Y, Zhang X, Wei X, Zhang Y, Wang D, Huang J, Zhu K, Peng G, Sun B. Aberrant serotonergic signaling contributes to the hyperexcitability of CA1 pyramidal neurons in a mouse model of Alzheimer's disease. Cell Rep 2023; 42:112152. [PMID: 36821438 DOI: 10.1016/j.celrep.2023.112152] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 11/29/2022] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Hyperactivity of pyramidal neurons (PNs) in CA1 is an early event in Alzheimer's disease. However, factors accounting for the hyperactivity of CA1 PNs remain to be completely investigated. In the present study, we report that the serotonergic signaling is abnormal in the hippocampus of hAPP-J20 mice. Interestingly, chemogenetic activation of serotonin (5-hydroxytryptamine; 5-HT) neurons in the median raphe nucleus (MRN) attenuates the activity of CA1 PNs in hAPP-J20 mice by regulating the intrinsic properties or inhibitory synaptic transmission of CA1 PNs through 5-HT3aR and/or 5-HT1aR. Furthermore, activating MRN 5-HT neurons improves memory in hAPP-J20 mice, and this effect is mediated by 5-HT3aR and 5-HT1aR. Direct activation of 5-HT3aR and 5-HT1aR with their selective agonists also improves the memory of hAPP-J20 mice. Together, we identify the impaired 5-HT/5-HT3aR and/or 5-HT/5-HT1aR signaling as pathways contributing to the hyperexcitability of CA1 PNs and the impaired cognition in hAPP-J20 mice.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurobiology and Department of Anesthesiology, the Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yufei Mei
- Department of Neurobiology and Department of Anesthesiology, the Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China; Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei 430081, China.
| | - Xiaoqin Zhang
- Department of Physiology and Pharmacology, Medical School of Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xiaojie Wei
- Department of Neurobiology and Department of Anesthesiology, the Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yiping Zhang
- Department of Neurobiology and Department of Anesthesiology, the Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Dongpi Wang
- Department of Anesthesiology, The Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310003, China
| | - Jinjin Huang
- Department of Anesthesiology, The Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310003, China
| | - Keqing Zhu
- National Human Brain Bank for Health and Disease and Department of Neurology in Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Binggui Sun
- Department of Neurobiology and Department of Anesthesiology, the Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310058, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
25
|
Niraula S, Doderer JJ, Indulkar S, Berry KP, Hauser WL, L'Esperance OJ, Deng JZ, Keeter G, Rouse AG, Subramanian J. Excitation-inhibition imbalance disrupts visual familiarity in amyloid and non-pathology conditions. Cell Rep 2023; 42:111946. [PMID: 36640331 PMCID: PMC9939293 DOI: 10.1016/j.celrep.2022.111946] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/14/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
Neuronal hyperactivity induces memory deficits in Alzheimer's disease. However, how hyperactivity disrupts memory is unclear. Using in vivo synaptic imaging in the mouse visual cortex, we show that structural excitatory-inhibitory synapse imbalance in the apical dendrites favors hyperactivity in early amyloidosis. Consistent with this, natural images elicit neuronal hyperactivity in these mice. Compensatory changes that maintain activity homeostasis disrupt functional connectivity and increase population sparseness such that a small fraction of neurons dominates population activity. These properties reduce the selectivity of neural response to natural images and render visual recognition memory vulnerable to interference. Deprivation of non-specific visual experiences improves the neural representation and behavioral expression of visual familiarity. In contrast, in non-pathological conditions, deprivation of non-specific visual experiences induces disinhibition, increases excitability, and disrupts visual familiarity. We show that disrupted familiarity occurs when the fraction of high-responsive neurons and the persistence of neural representation of a memory-associated stimulus are not constrained.
Collapse
Affiliation(s)
- Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Julia J Doderer
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Shreya Indulkar
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Kalen P Berry
- Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - William L Hauser
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Oliver J L'Esperance
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Jasmine Z Deng
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Griffin Keeter
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Adam G Rouse
- Department of Neurosurgery, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
26
|
Li Z, Cao Y, Pei H, Ma L, Yang Y, Li H. The contribution of mitochondria-associated endoplasmic reticulum membranes (MAMs) dysfunction in Alzheimer's disease and the potential countermeasure. Front Neurosci 2023; 17:1158204. [PMID: 36960176 PMCID: PMC10027904 DOI: 10.3389/fnins.2023.1158204] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. There are many studies targeting extracellular deposits of amyloid β-peptide (Aβ) and intracellular neurofibrillary tangles (NFTs), however, there are no effective treatments to halt the progression. Mitochondria-associated endoplasmic reticulum membranes (MAMs) have long been found to be associated with various pathogenesis hypotheses of AD, such as Aβ deposition, mitochondrial dysfunction, and calcium homeostasis. However, there is a lack of literature summarizing recent advances in the mechanism and treatment studies. Accordingly, this article reviews the latest research involving the roles of MAM structure and tethering proteins in the pathogenesis of AD and summarizes potential strategies targeting MAMs to dissect treatment perspectives for AD.
Collapse
Affiliation(s)
- Zehui Li
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Pei
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yang Yang
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yang Yang,
| | - Hao Li
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Hao Li,
| |
Collapse
|
27
|
Mustaly-Kalimi S, Gallegos W, Marr RA, Gilman-Sachs A, Peterson DA, Sekler I, Stutzmann GE. Protein mishandling and impaired lysosomal proteolysis generated through calcium dysregulation in Alzheimer's disease. Proc Natl Acad Sci U S A 2022; 119:e2211999119. [PMID: 36442130 PMCID: PMC9894236 DOI: 10.1073/pnas.2211999119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022] Open
Abstract
Impairments in neural lysosomal- and autophagic-mediated degradation of cellular debris contribute to neuritic dystrophy and synaptic loss. While these are well-characterized features of neurodegenerative disorders such as Alzheimer's disease (AD), the upstream cellular processes driving deficits in pathogenic protein mishandling are less understood. Using a series of fluorescent biosensors and optical imaging in model cells, AD mouse models and human neurons derived from AD patients, we reveal a previously undescribed cellular signaling cascade underlying protein mishandling mediated by intracellular calcium dysregulation, an early component of AD pathogenesis. Increased Ca2+ release via the endoplasmic reticulum (ER)-resident ryanodine receptor (RyR) is associated with reduced expression of the lysosome proton pump vacuolar-ATPase (vATPase) subunits (V1B2 and V0a1), resulting in lysosome deacidification and disrupted proteolytic activity in AD mouse models and human-induced neurons (HiN). As a result of impaired lysosome digestive capacity, mature autophagosomes with hyperphosphorylated tau accumulated in AD murine neurons and AD HiN, exacerbating proteinopathy. Normalizing AD-associated aberrant RyR-Ca2+ signaling with the negative allosteric modulator, dantrolene (Ryanodex), restored vATPase levels, lysosomal acidification and proteolytic activity, and autophagic clearance of intracellular protein aggregates in AD neurons. These results highlight that prior to overt AD histopathology or cognitive deficits, aberrant upstream Ca2+ signaling disrupts lysosomal acidification and contributes to pathological accumulation of intracellular protein aggregates. Importantly, this is demonstrated in animal models of AD, and in human iPSC-derived neurons from AD patients. Furthermore, pharmacological suppression of RyR-Ca2+ release rescued proteolytic function, revealing a target for therapeutic intervention that has demonstrated effects in clinically-relevant assays.
Collapse
Affiliation(s)
- Sarah Mustaly-Kalimi
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Wacey Gallegos
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Robert A. Marr
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Alice Gilman-Sachs
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University of Medicine and Science, Immunology, and Infection, North Chicago, IL60064
| | - Daniel A. Peterson
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Science and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva84105, Israel
| | - Grace E. Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| |
Collapse
|
28
|
Berkowitz BA, Podolsky RH, Childers KL, Roberts R, Katz R, Waseem R, Robbings BM, Hass DT, Hurley JB, Sweet IR, Goodman C, Qian H, Alvisio B, Heaps S. Transducin-Deficient Rod Photoreceptors Evaluated With Optical Coherence Tomography and Oxygen Consumption Rate Energy Biomarkers. Invest Ophthalmol Vis Sci 2022; 63:22. [PMID: 36576748 PMCID: PMC9804021 DOI: 10.1167/iovs.63.13.22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Purpose To test the hypothesis that rod energy biomarkers in light and dark are similar in mice without functional rod transducin (Gnat1rd17). Methods Gnat1rd17 and wildtype (WT) mice were studied in canonically low energy demand (light) and high energy demand (dark) conditions. We measured rod inner segment ellipsoid zone (ISez) profile shape, external limiting membrane-retinal pigment epithelium (ELM-RPE) thickness, and magnitude of a hyporeflective band (HB) intensity dip located between photoreceptor tips and apical RPE; antioxidants were given in a subset of mice. Oxygen consumption rate (OCR) and visual performance indexes were also measured. Results The lower energy demand expected in light-adapted wildtype retinas was associated with an elongated ISez, thicker ELM-RPE, and higher HB magnitude, and lower OCR compared to high energy demand conditions in the dark. Gnat1rd17 mice showed a wildtype-like ISez profile shape at 20 minutes of light that became rounder at 60 minutes; at both times, ELM-RPE was smaller than wildtype values, and the HB magnitude was unmeasurable. OCR was higher than in the dark. Light-adapted Gnat1rd17 mice biomarkers were unaffected by anti-oxidants. Gnat1rd17 mice showed modest outer nuclear layer thinning and no reduction in visual performance indexes. Conclusions Light-stimulated changes in all biomarkers in WT mice are consistent with the established light-induced decrease in net energy demand. In contrast, biomarker changes in Gnat1rd17 mice raise the possibility that light increases net energy demand in the absence of rod phototransduction.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Robert H Podolsky
- Biostatistics and Study Methodology, Children's National Hospital, Silver Spring, Maryland, United States
| | - Karen Lins Childers
- Beaumont Research Institute, Beaumont Health, Royal Oak, Michigan, United States
| | - Robin Roberts
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Ryan Katz
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Rida Waseem
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Brian M Robbings
- Department of Biochemistry, Department of Ophthalmology, University of Washington, Seattle, Washington, United States.,Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - Daniel T Hass
- Department of Biochemistry, Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| | - James B Hurley
- Department of Biochemistry, Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| | - Ian R Sweet
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - Cole Goodman
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Bruno Alvisio
- OSIO Bioinformatics Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Sam Heaps
- OSIO Bioinformatics Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
29
|
Yao J, Chen SRW. R-carvedilol, a potential new therapy for Alzheimer's disease. Front Pharmacol 2022; 13:1062495. [PMID: 36532759 PMCID: PMC9756136 DOI: 10.3389/fphar.2022.1062495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
Abstract
For decades, the amyloid cascade hypothesis has been the leading hypothesis in studying Alzheimer's disease (AD) pathology and drug development. However, a growing body of evidence indicates that simply removing amyloid plaques may not significantly affect AD progression. Alternatively, it has been proposed that AD progression is driven by increased neuronal excitability. Consistent with this alternative hypothesis, recent studies showed that pharmacologically limiting ryanodine receptor 2 (RyR2) open time with the R-carvedilol enantiomer prevented and reversed neuronal hyperactivity, memory impairment, and neuron loss in AD mouse models without affecting the accumulation of ß-amyloid (Aβ). These data indicate that R-carvedilol could be a potential new therapy for AD.
Collapse
Affiliation(s)
- Jinjing Yao
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada,Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,*Correspondence: Jinjing Yao, ; S. R. Wayne Chen,
| | - S. R. Wayne Chen
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada,Department of Physiology and Pharmacology, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,*Correspondence: Jinjing Yao, ; S. R. Wayne Chen,
| |
Collapse
|
30
|
Yao J, Liu Y, Sun B, Zhan X, Estillore JP, Turner RW, Chen SRW. Increased RyR2 open probability induces neuronal hyperactivity and memory loss with or without Alzheimer's disease-causing gene mutations. Alzheimers Dement 2022; 18:2088-2098. [PMID: 34985200 DOI: 10.1002/alz.12543] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/01/2021] [Accepted: 10/25/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Neuronal hyperactivity is an early neuronal defect commonly observed in familial and sporadic Alzheimer's disease (AD), but the underlying mechanisms are unclear. METHODS We employed a ryanodine receptor 2 (RyR2) mutant mouse model harboring the R4496C+/- mutation that markedly increases the channel's open probability (Po) to determine the impact of increased RyR2 activity in neuronal function without AD gene mutations. RESULTS Genetically increasing RyR2 Po induced neuronal hyperactivity in vivo in anesthetized and awake mice. Increased RyR2 Po induced hyperactive behaviors, impaired learning and memory, defective dendritic spines, and neuronal cell death. Increased RyR2 Po exacerbated the onset of neuronal hyperexcitability and learning and memory impairments in 5xFAD mice. DISCUSSION Increased RyR2 Po exacerbates the onset of familial AD-associated neuronal dysfunction, and induces AD-like defects in the absence of AD-causing gene mutations, suggesting that RyR2-associated neuronal hyperactivity represents a common target for combating AD with or without AD gene mutations.
Collapse
Affiliation(s)
- Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yajing Liu
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiaoqin Zhan
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - John Paul Estillore
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ray W Turner
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
31
|
Zhan X, Drummond-Main C, Greening D, Yao J, Chen SWR, Appendino JP, Au PYB, Turner RW. Cannabidiol counters the effects of a dominant-negative pathogenic Kv7.2 variant. iScience 2022; 25:105092. [PMID: 36157585 PMCID: PMC9490039 DOI: 10.1016/j.isci.2022.105092] [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: 06/02/2022] [Revised: 08/17/2022] [Accepted: 09/02/2022] [Indexed: 11/04/2022] Open
Abstract
Epilepsy and neurodevelopmental disorders can arise from pathogenic variants of KCNQ (Kv7) channels. A patient with developmental and epileptic encephalopathy exhibited an in-frame deletion of histidine 260 on Kv7.2. Coexpression of Kv7.2 mutant (mut) subunits with Kv7.3 invoked a decrease in current density, a depolarizing shift in voltage for activation, and a decrease in membrane conductance. Biotinylation revealed an increased level of surface Kv7.2mut compared to Kv7.3 with no change in total membrane protein expression. Super-resolution and FRET imaging confirmed heteromeric channel formation and a higher expression density of Kv7.2mut. Cannabidiol (1 μM) offset the effects of Kv7.2mut by inducing a hyperpolarizing shift in voltage for activation independent of CB1 or CB2 receptors. These data reveal that the ability for cannabidiol to reduce the effects of a pathogenic Kv7.2 variant supports its use as a potential therapeutic to reduce seizure activity. A patient with epileptic encephalopathy exhibits a Kv7.2 deletion at H260 (Kv7.2mut) Kv7.2mut shows increased expression at the membrane compared to Kv7.3 Kv7.2mut acts in a dominant-negative manner to reduce Kv7 conductance Cannabidiol acts on Kv7.x activation voltage to offset the effects of Kv7.2mut
Collapse
Affiliation(s)
- Xiaoqin Zhan
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Chris Drummond-Main
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Dylan Greening
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Jinjing Yao
- Libin Cardiovascular Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - S W R Chen
- Libin Cardiovascular Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - J P Appendino
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Pediatric Department, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - P Y Billie Au
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Medical Genetics, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Ray W Turner
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
32
|
Ohline SM, Liu X, Ibrahim MF, Mockett BM, Empson RM, Abraham WC, Iremonger KJ, Jones PP. Altered membrane properties but unchanged intrinsic excitability and spontaneous postsynaptic currents in an aged APPswe/PS1dE9 model of Alzheimer’s disease. Front Cell Neurosci 2022; 16:958876. [PMID: 36090787 PMCID: PMC9459330 DOI: 10.3389/fncel.2022.958876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Neuronal hyperexcitability in Alzheimer’s disease (AD) models is thought to either contribute to the formation of amyloid beta plaques or result from their formation. Neuronal hyperexcitability has been shown in the cerebral cortex of the widely used young APPswe/PS1dE9 mice, which have accelerated plaque formation. However, it is currently unclear if hyperexcitability also occurs in CA1 hippocampal neurons of aged animals in this model. In the present work, we have compared intrinsic excitability and spontaneous synaptic inputs from CA1 pyramidal cells of 8-month-old APPswe/PS1dE9 and wildtype control mice. We find no change in intrinsic excitability or spontaneous postsynaptic currents (PSCs) between groups. We did, however, find a reduced input resistance and an increase in hyperpolarization-activated sag current. These results are consistent with findings from other aged AD model mice, including the widely used 5xFAD and 3xTg. Together these results suggest that neuronal hyperexcitability is not a consistent feature of all AD mouse models, particularly at advanced ages.
Collapse
Affiliation(s)
- Shane M. Ohline
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- HeartOtago, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- *Correspondence: Shane M. Ohline,
| | - Xinhuai Liu
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Mohamed F. Ibrahim
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, United Kingdom
| | - Bruce M. Mockett
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Ruth M. Empson
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Wickliffe C. Abraham
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Karl J. Iremonger
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Peter P. Jones
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- HeartOtago, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Peter P. Jones,
| |
Collapse
|
33
|
Zou HY, Guo L, Zhang B, Chen S, Wu XR, Liu XD, Xu XY, Li BY, Chen S, Xu NJ, Sun S. Aberrant miR-339-5p/neuronatin signaling causes prodromal neuronal calcium dyshomeostasis in mutant presenilin mice. J Clin Invest 2022; 132:e149160. [PMID: 35426376 PMCID: PMC9012292 DOI: 10.1172/jci149160] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 02/23/2022] [Indexed: 12/22/2022] Open
Abstract
Mushroom spine loss and calcium dyshomeostasis are early hallmark events of age-related neurodegeneration, such as Alzheimer's disease (AD), that are connected with neuronal hyperactivity in early pathology of cognitive brain areas. However, it remains elusive how these key events are triggered at the molecular level for the neuronal abnormality that occurs at the initial stage of disease. Here, we identify downregulated miR-339-5p and its upregulated target protein, neuronatin (Nnat), in cortex neurons from the presenilin-1 M146V knockin (PSEN1-M146V KI) mouse model of familial AD (FAD). Inhibition of miR-339-5p or overexpression of Nnat recapitulates spine loss and endoplasmic reticulum calcium overload in cortical neurons with the PSEN1 mutation. Conversely, either overexpression of miR-339-5p or knockdown of Nnat restores spine morphogenesis and calcium homeostasis. We used fiber photometry recording during the object-cognitive process to further demonstrate that the PSEN1 mutant causes defective habituation in neuronal reaction in the retrosplenial cortex and that this can be rescued by restoring the miR-339-5p/Nnat pathway. Our findings thus reveal crucial roles of the miR-339-5p/Nnat pathway in FAD that may serve as potential diagnostic and therapeutic targets for early pathogenesis.
Collapse
Affiliation(s)
- Hao-Yu Zou
- Department of Neurology and Institute of Neurology, Ruijin Hospital
| | - Lin Guo
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, and
| | - Bei Zhang
- Department of Neurology and Institute of Neurology, Ruijin Hospital
| | - Si Chen
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, and
| | - Xin-Rong Wu
- Department of Neurology and Institute of Neurology, Ruijin Hospital
| | - Xian-Dong Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, and
| | - Xin-Yu Xu
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, and
| | - Bin-Yin Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital
| | - Nan-Jie Xu
- Research Center of Translational Medicine, Shanghai Children’s Hospital, Department of Anatomy and Physiology, and
- Shanghai Key Laboratory of Reproductive Medicine, and
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suya Sun
- Department of Neurology and Institute of Neurology, Ruijin Hospital
| |
Collapse
|
34
|
Hiess F, Yao J, Song Z, Sun B, Zhang Z, Huang J, Chen L, Institoris A, Estillore JP, Wang R, Ter Keurs HEDJ, Stys PK, Gordon GR, Zamponi GW, Ganguly A, Chen SRW. Subcellular localization of hippocampal ryanodine receptor 2 and its role in neuronal excitability and memory. Commun Biol 2022; 5:183. [PMID: 35233070 PMCID: PMC8888588 DOI: 10.1038/s42003-022-03124-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/01/2022] [Indexed: 11/09/2022] Open
Abstract
Ryanodine receptor 2 (RyR2) is abundantly expressed in the heart and brain. Mutations in RyR2 are associated with both cardiac arrhythmias and intellectual disability. While the mechanisms of RyR2-linked arrhythmias are well characterized, little is known about the mechanism underlying RyR2-associated intellectual disability. Here, we employed a mouse model expressing a green fluorescent protein (GFP)-tagged RyR2 and a specific GFP probe to determine the subcellular localization of RyR2 in hippocampus. GFP-RyR2 was predominantly detected in the soma and dendrites, but not the dendritic spines of CA1 pyramidal neurons or dentate gyrus granular neurons. GFP-RyR2 was also detected within the mossy fibers in the stratum lucidum of CA3, but not in the presynaptic terminals of CA1 neurons. An arrhythmogenic RyR2-R4496C+/− mutation downregulated the A-type K+ current and increased membrane excitability, but had little effect on the afterhyperpolarization current or presynaptic facilitation of CA1 neurons. The RyR2-R4496C+/− mutation also impaired hippocampal long-term potentiation, learning, and memory. These data reveal the precise subcellular distribution of hippocampal RyR2 and its important role in neuronal excitability, learning, and memory. A mouse model containing a GFP-tagged ryanodine receptor 2 (RyR2) has shed light on the precise subcellular localization of hippocampal RyR2 and mechanisms underlying neuronal excitability, learning, and memory.
Collapse
Affiliation(s)
- Florian Hiess
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Zhenpeng Song
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Zizhen Zhang
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Junting Huang
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Lina Chen
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - John Paul Estillore
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Henk E D J Ter Keurs
- Libin Cardiovascular Institute, Department of Cardiovascular Science, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Peter K Stys
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.,Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Gerald W Zamponi
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Anutosh Ganguly
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, T2N 4N1, Canada. .,Hotchkiss Brain Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
35
|
Jiang H, Wang G, Gu J, Xiao Y, Wang P, Huang X, Sha H, Wang Z, Ma Q. Resveratrol inhibits the expression of RYR2 and is a potential treatment for pancreatic cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:315-324. [PMID: 35044499 DOI: 10.1007/s00210-022-02203-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/08/2022] [Indexed: 10/19/2022]
Abstract
Resveratrol is a polyphonous natural compound that has cardioprotective, anticancer, and anti-inflammatory properties. Studies have proved that resveratrol (RES) inhibits cancer cell proliferation, migration, and invasion and promotes apoptosis. Elevated expression of ryanodine receptor type 2 (RYR2) may participate in the pathway responsible for calcium metabolism as well as anti-apoptosis and anti-autophagy events in malignant tumor cells. However, the underlying molecular mechanisms of RES anticancer effects with RYR2 are not completely understood in pancreatic cancer. The aim of the present study was tantamount to study the effect of RES in human pancreatic cancer and investigate the underlying mechanisms of RES. We found that RES inhibits proliferation, migration, and invasion and suppresses RYR2 expression in pancreatic cancer cells. In addition, RYR2 knockdown impedes the proliferation, migration, and invasiveness of pancreatic cancer cells. RYR2 knockdown can also increase PTEN expression, while increased RYR2 expression can inhibit PTEN expression. Moreover, RES can upregulate PTEN expression. Taken together, these results indicate that RES could play an antitumor role by decreasing RYR2 expression.
Collapse
Affiliation(s)
- He Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Guotai Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China.,Department of Hepatobiliary Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Jingtao Gu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ying Xiao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Pengli Wang
- Department of Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Xin Huang
- Department of General Surgery, Xi'an Central Hospital, Xi'an, 710003, China
| | - Huanchen Sha
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
36
|
Prista von Bonhorst F, Gall D, Dupont G. Impact of β-Amyloids Induced Disruption of Ca2+ Homeostasis in a Simple Model of Neuronal Activity. Cells 2022; 11:cells11040615. [PMID: 35203266 PMCID: PMC8869902 DOI: 10.3390/cells11040615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 01/25/2023] Open
Abstract
Alzheimer’s disease is characterized by a marked dysregulation of intracellular Ca2+ homeostasis. In particular, toxic β-amyloids (Aβ) perturb the activities of numerous Ca2+ transporters or channels. Because of the tight coupling between Ca2+ dynamics and the membrane electrical activity, such perturbations are also expected to affect neuronal excitability. We used mathematical modeling to systematically investigate the effects of changing the activities of the various targets of Aβ peptides reported in the literature on calcium dynamics and neuronal excitability. We found that the evolution of Ca2+ concentration just below the plasma membrane is regulated by the exchanges with the extracellular medium, and is practically independent from the Ca2+ exchanges with the endoplasmic reticulum. Thus, disruptions of Ca2+ homeostasis interfering with signaling do not affect the electrical properties of the neurons at the single cell level. In contrast, the model predicts that by affecting the activities of L-type Ca2+ channels or Ca2+-activated K+ channels, Aβ peptides promote neuronal hyperexcitability. On the contrary, they induce hypo-excitability when acting on the plasma membrane Ca2+ ATPases. Finally, the presence of pores of amyloids in the plasma membrane can induce hypo- or hyperexcitability, depending on the conditions. These modeling conclusions should help with analyzing experimental observations in which Aβ peptides interfere at several levels with Ca2+ signaling and neuronal activity.
Collapse
Affiliation(s)
- Francisco Prista von Bonhorst
- Unit of Theoretical Chronobiology, Faculté des Sciences (CP231), Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium;
| | - David Gall
- Research Laboratory in Human Reproduction, Faculté de Médecine (CP636), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
| | - Geneviève Dupont
- Unit of Theoretical Chronobiology, Faculté des Sciences (CP231), Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium;
- Correspondence:
| |
Collapse
|
37
|
Wang T, Chen Y, Zou Y, Pang Y, He X, Chen Y, Liu Y, Feng W, Zhang Y, Li Q, Shi J, Ding F, Marshall C, Gao J, Xiao M. Locomotor Hyperactivity in the Early-Stage Alzheimer’s Disease-like Pathology of APP/PS1 Mice: Associated with Impaired Polarization of Astrocyte Aquaporin 4. Aging Dis 2022; 13:1504-1522. [PMID: 36186142 PMCID: PMC9466968 DOI: 10.14336/ad.2022.0219] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/19/2022] [Indexed: 12/21/2022] Open
Abstract
Non-cognitive behavioral and psychological symptoms often occur in Alzheimer's disease (AD) patients and mouse models, although the exact neuropathological mechanism remains elusive. Here, we report hyperactivity with significant inter-individual variability in 4-month-old APP/PS1 mice. Pathological analysis revealed that intraneuronal accumulation of amyloid-β (Aβ), c-Fos expression in glutamatergic neurons and activation of astrocytes were more evident in the frontal motor cortex of hyperactive APP/PS1 mice, compared to those with normal activity. Moreover, the hyperactive phenotype was associated with mislocalization of perivascular aquaporin 4 (AQP4) and glymphatic transport impairment. Deletion of the AQP4 gene increased hyperactivity, intraneuronal Aβ load and glutamatergic neuron activation, but did not influence working memory or anxiety-like behaviors of 4-month-old APP/PS1 mice. Together, these results demonstrate that AQP4 mislocalization or deficiency leads to increased intraneuronal Aβ load and neuronal hyperactivity in the motor cortex, which in turn causes locomotor over-activity during the early pathophysiology of APP/PS1 mice. Therefore, improving AQP4 mediated glymphatic clearance may offer a new strategy for early intervention of hyperactivity in the prodromal phase of AD.
Collapse
Affiliation(s)
- Tianqi Wang
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yan Chen
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Ying Zou
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Yingting Pang
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiaoxin He
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yali Chen
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yun Liu
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Weixi Feng
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yanli Zhang
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Qian Li
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Jingping Shi
- Department of Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Fengfei Ding
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Charles Marshall
- College of Health Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY 41701, USA
| | - Junying Gao
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Correspondence should be addressed to: Dr. Ming Xiao (E-mail: ) or Dr. Junying Gao (), Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Correspondence should be addressed to: Dr. Ming Xiao (E-mail: ) or Dr. Junying Gao (), Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| |
Collapse
|
38
|
Sleiman Y, Lacampagne A, Meli AC. "Ryanopathies" and RyR2 dysfunctions: can we further decipher them using in vitro human disease models? Cell Death Dis 2021; 12:1041. [PMID: 34725342 PMCID: PMC8560800 DOI: 10.1038/s41419-021-04337-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 12/23/2022]
Abstract
The regulation of intracellular calcium (Ca2+) homeostasis is fundamental to maintain normal functions in many cell types. The ryanodine receptor (RyR), the largest intracellular calcium release channel located on the sarco/endoplasmic reticulum (SR/ER), plays a key role in the intracellular Ca2+ handling. Abnormal type 2 ryanodine receptor (RyR2) function, associated to mutations (ryanopathies) or pathological remodeling, has been reported, not only in cardiac diseases, but also in neuronal and pancreatic disorders. While animal models and in vitro studies provided valuable contributions to our knowledge on RyR2 dysfunctions, the human cell models derived from patients’ cells offer new hope for improving our understanding of human clinical diseases and enrich the development of great medical advances. We here discuss the current knowledge on RyR2 dysfunctions associated with mutations and post-translational remodeling. We then reviewed the novel human cellular technologies allowing the correlation of patient’s genome with their cellular environment and providing approaches for personalized RyR-targeted therapeutics.
Collapse
Affiliation(s)
- Yvonne Sleiman
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Albano C Meli
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.
| |
Collapse
|
39
|
Liu Y, Yao J, Song Z, Guo W, Sun B, Wei J, Estillore JP, Back TG, Chen SRW. Limiting RyR2 open time prevents Alzheimer's disease-related deficits in the 3xTG-AD mouse model. J Neurosci Res 2021; 99:2906-2921. [PMID: 34352124 DOI: 10.1002/jnr.24936] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/11/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022]
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) progression is driven by a vicious cycle of soluble β-amyloid (Aβ)-induced neuronal hyperactivity. Thus, breaking this vicious cycle by suppressing neuronal hyperactivity may represent a logical approach to stopping AD progression. In support of this, we have recently shown that genetically and pharmacologically limiting ryanodine receptor 2 (RyR2) open time prevented neuronal hyperactivity, memory impairment, dendritic spine loss, and neuronal cell death in a rapid, early onset AD mouse model (5xFAD). Here, we assessed the impact of limiting RyR2 open time on AD-related deficits in a relatively late occurring, slow developing AD mouse model (3xTG-AD) that bears more resemblance (compared to 5xFAD) to that of human AD. Using behavioral tests, long-term potentiation recordings, and Golgi and Nissl staining, we found that the RyR2-E4872Q mutation, which markedly shortens the open duration of the RyR2 channel, prevented learning and memory impairment, defective long-term potentiation, dendritic spine loss, and neuronal cell death in the 3xTG-AD mice. Furthermore, pharmacologically shortening the RyR2 open time with R-carvedilol rescued these AD-related deficits in 3xTG mice. Therefore, limiting RyR2 open time may offer a promising, neuronal hyperactivity-targeted anti-AD strategy.
Collapse
Affiliation(s)
- Yajing Liu
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinjing Yao
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Zhenpeng Song
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Wenting Guo
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Bo Sun
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jinhong Wei
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - John Paul Estillore
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Thomas G Back
- Department of Chemistry, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
40
|
Sun B, Yao J, Chen AW, Estillore JP, Wang R, Back TG, Chen SRW. Genetically and pharmacologically limiting RyR2 open time prevents neuronal hyperactivity of hippocampal CA1 neurons in brain slices of 5xFAD mice. Neurosci Lett 2021; 758:136011. [PMID: 34090936 DOI: 10.1016/j.neulet.2021.136011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/19/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022]
Abstract
Neuronal hyperactivity is an early, common manifestation of Alzheimer's disease (AD), and is believed to drive AD progression. Neuronal hyperactivity in the form of baseline activity (or spontaneous Ca2+ transients) has consistently been demonstrated in mouse models of AD using two-photon in vivo Ca2+ imaging of cortical or hippocampal neurons in anesthetized animals. Notably, these AD-related spontaneous Ca2+ transients were hardly detected in acute hippocampal slices, probably due to neuronal damage during brain slicing. To better preserve neuronal activity, we employed the N-methyl-D-glucamine (NMDG) protective brain slicing protocol. We performed confocal in vitro Ca2+ imaging of hippocampal CA1 neurons in optimized hippocampal slices. Consistent with previous in vivo studies, our in vitro studies using optimized brain slices also showed that limiting the open duration of the ryanodine receptor 2 (RyR2) by the RyR2 mutation E4872Q or by the R-carvedilol enantiomer prevented and rescued neuronal hyperactivity of hippocampal CA1 neurons from 5xFAD mice. Thus, genetically and pharmacologically limiting RyR2 open time prevented and rescued AD-related neuronal hyperactivity in vitro in optimized brain slices in the absence of anesthetics' influence. Our data also suggest that the NMDG protective brain slicing preparation offers an alternative means to study neuronal hyperactivity of various cell types in different brain regions, especially in regions that are not readily accessible to two-photon in vivo Ca2+ imaging.
Collapse
Affiliation(s)
- Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Alexander W Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - John Paul Estillore
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Thomas G Back
- Department of Chemistry, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
41
|
Guo W, Wei J, Estillore JP, Zhang L, Wang R, Sun B, Chen SRW. RyR2 disease mutations at the C-terminal domain intersubunit interface alter closed-state stability and channel activation. J Biol Chem 2021; 297:100808. [PMID: 34022226 PMCID: PMC8214192 DOI: 10.1016/j.jbc.2021.100808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 11/19/2022] Open
Abstract
Ryanodine receptors (RyRs) are ion channels that mediate the release of Ca2+ from the sarcoplasmic reticulum/endoplasmic reticulum, mutations of which are implicated in a number of human diseases. The adjacent C-terminal domains (CTDs) of cardiac RyR (RyR2) interact with each other to form a ring-like tetrameric structure with the intersubunit interface undergoing dynamic changes during channel gating. This mobile CTD intersubunit interface harbors many disease-associated mutations. However, the mechanisms of action of these mutations and the role of CTD in channel function are not well understood. Here, we assessed the impact of CTD disease-associated mutations P4902S, P4902L, E4950K, and G4955E on Ca2+− and caffeine-mediated activation of RyR2. The G4955E mutation dramatically increased both the Ca2+-independent basal activity and Ca2+-dependent activation of [3H]ryanodine binding to RyR2. The P4902S and E4950K mutations also increased Ca2+ activation but had no effect on the basal activity of RyR2. All four disease mutations increased caffeine-mediated activation of RyR2 and reduced the threshold for activation and termination of spontaneous Ca2+ release. G4955D dramatically increased the basal activity of RyR2, whereas G4955K mutation markedly suppressed channel activity. Similarly, substitution of P4902 with a negatively charged residue (P4902D), but not a positively charged residue (P4902K), also dramatically increased the basal activity of RyR2. These data suggest that electrostatic interactions are involved in stabilizing the CTD intersubunit interface and that the G4955E disease mutation disrupts this interface, and thus the stability of the closed state. Our studies shed new insights into the mechanisms of action of RyR2 CTD disease mutations.
Collapse
Affiliation(s)
- Wenting Guo
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Jinhong Wei
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - John Paul Estillore
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Lin Zhang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; Medical School, Kunming University of Science and Technology, Kunming, China.
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
42
|
McCarty MF, DiNicolantonio JJ, Lerner A. A Fundamental Role for Oxidants and Intracellular Calcium Signals in Alzheimer's Pathogenesis-And How a Comprehensive Antioxidant Strategy May Aid Prevention of This Disorder. Int J Mol Sci 2021; 22:2140. [PMID: 33669995 PMCID: PMC7926325 DOI: 10.3390/ijms22042140] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress and increased cytoplasmic calcium are key mediators of the detrimental effects on neuronal function and survival in Alzheimer's disease (AD). Pathways whereby these perturbations arise, and then prevent dendritic spine formation, promote tau hyperphosphorylation, further amplify amyloid β generation, and induce neuronal apoptosis, are described. A comprehensive program of nutraceutical supplementation, comprised of the NADPH oxidase inhibitor phycocyanobilin, phase two inducers, the mitochondrial antioxidant astaxanthin, and the glutathione precursor N-acetylcysteine, may have important potential for antagonizing the toxic effects of amyloid β on neurons and thereby aiding prevention of AD. Moreover, nutraceutical antioxidant strategies may oppose the adverse impact of amyloid β oligomers on astrocyte clearance of glutamate, and on the ability of brain capillaries to export amyloid β monomers/oligomers from the brain. Antioxidants, docosahexaenoic acid (DHA), and vitamin D, have potential for suppressing microglial production of interleukin-1β, which potentiates the neurotoxicity of amyloid β. Epidemiology suggests that a health-promoting lifestyle, incorporating a prudent diet, regular vigorous exercise, and other feasible measures, can cut the high risk for AD among the elderly by up to 60%. Conceivably, complementing such lifestyle measures with long-term adherence to the sort of nutraceutical regimen outlined here may drive down risk for AD even further.
Collapse
Affiliation(s)
| | | | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer 5262000, Israel
| |
Collapse
|
43
|
Stutzmann GE. RyR2 calcium channels in the spotlight-I'm ready for my close up, Dr. Alzheimer! Cell Calcium 2021; 94:102342. [PMID: 33444912 DOI: 10.1016/j.ceca.2020.102342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 12/26/2020] [Accepted: 12/26/2020] [Indexed: 02/03/2023]
Abstract
While various hypotheses surrounding the etiology of Alzheimer's disease (AD) have waxed and waned over the years, the calcium hypothesis of aging [1] has maintained its steady trajectory since the early 1990's, albeit often as the understudy. Here, Yao et al., [2] further implicate intracellular calcium dysregulation in AD pathogenesis, and focus the spotlight on the elusive ryanodine receptor-2 isoform.
Collapse
Affiliation(s)
- Grace E Stutzmann
- Rosalind Franklin University of Medicine and Science, Chicago Medical School, Center for Neurodegenerative Disease and Therapeutics, 3333 Green Bay Rd., North Chicago, IL, 60064, United States.
| |
Collapse
|
44
|
Ca 2+ Dyshomeostasis Disrupts Neuronal and Synaptic Function in Alzheimer's Disease. Cells 2020; 9:cells9122655. [PMID: 33321866 PMCID: PMC7763805 DOI: 10.3390/cells9122655] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Ca2+ homeostasis is essential for multiple neuronal functions and thus, Ca2+ dyshomeostasis can lead to widespread impairment of cellular and synaptic signaling, subsequently contributing to dementia and Alzheimer's disease (AD). While numerous studies implicate Ca2+ mishandling in AD, the cellular basis for loss of cognitive function remains under investigation. The process of synaptic degradation and degeneration in AD is slow, and constitutes a series of maladaptive processes each contributing to a further destabilization of the Ca2+ homeostatic machinery. Ca2+ homeostasis involves precise maintenance of cytosolic Ca2+ levels, despite extracellular influx via multiple synaptic Ca2+ channels, and intracellular release via organelles such as the endoplasmic reticulum (ER) via ryanodine receptor (RyRs) and IP3R, lysosomes via transient receptor potential mucolipin channel (TRPML) and two pore channel (TPC), and mitochondria via the permeability transition pore (PTP). Furthermore, functioning of these organelles relies upon regulated inter-organelle Ca2+ handling, with aberrant signaling resulting in synaptic dysfunction, protein mishandling, oxidative stress and defective bioenergetics, among other consequences consistent with AD. With few effective treatments currently available to mitigate AD, the past few years have seen a significant increase in the study of synaptic and cellular mechanisms as drivers of AD, including Ca2+ dyshomeostasis. Here, we detail some key findings and discuss implications for future AD treatments.
Collapse
|