1
|
Bussy A, Patel R, Parent O, Salaciak A, Bedford SA, Farzin S, Tullo S, Picard C, Villeneuve S, Poirier J, Breitner JC, Devenyi GA, Tardif CL, Chakravarty MM. Exploring morphological and microstructural signatures across the Alzheimer's spectrum and risk factors. Neurobiol Aging 2025; 149:1-18. [PMID: 39961166 DOI: 10.1016/j.neurobiolaging.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 03/15/2025]
Abstract
Neural alterations, including myelin degeneration and inflammation-related iron burden, may accompany early Alzheimer's disease (AD) pathophysiology. This study aims to identify multi-modal signatures associated with MRI-derived atrophy and quantitative MRI (qMRI) measures of myelin and iron in a unique dataset of 158 participants across the AD spectrum, including those without cognitive impairment, at familial risk for AD, with mild cognitive impairment, and with AD dementia. Our results revealed a brain pattern with decreased cortical thickness, indicating increased neuronal death, and compromised hippocampal integrity due to reduced myelin content. This pattern was associated with lifestyle factors such as smoking, high blood pressure, high cholesterol, and anxiety, as well as older age, AD progression, and APOE-ɛ4 carrier status. These findings underscore the value of qMRI metrics as a non-invasive tool, offering sensitivity to lifestyle-related modifiable risk factors and medical history, even in preclinical stages of AD.
Collapse
Affiliation(s)
- Aurélie Bussy
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Research Centre, 6875 Bd LaSalle CIC Building, Verdun, QC H4H 1R3, Canada; Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Room 302, Irving Ludmer Building, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada.
| | - Raihaan Patel
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Research Centre, 6875 Bd LaSalle CIC Building, Verdun, QC H4H 1R3, Canada; Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Department of Biomedical Engineering, McGill University, Duff Medical Building, 3775 Rue University Suite 316, Montreal, QC H3A 2B4, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Olivier Parent
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Research Centre, 6875 Bd LaSalle CIC Building, Verdun, QC H4H 1R3, Canada; Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Room 302, Irving Ludmer Building, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Alyssa Salaciak
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Research Centre, 6875 Bd LaSalle CIC Building, Verdun, QC H4H 1R3, Canada; Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Saashi A Bedford
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Research Centre, 6875 Bd LaSalle CIC Building, Verdun, QC H4H 1R3, Canada; Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Sarah Farzin
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Research Centre, 6875 Bd LaSalle CIC Building, Verdun, QC H4H 1R3, Canada; Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Stephanie Tullo
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Research Centre, 6875 Bd LaSalle CIC Building, Verdun, QC H4H 1R3, Canada; Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Room 302, Irving Ludmer Building, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Avenue West, Montreal, QC H3A 1A1, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, 3801 University St, Montreal, QC H3A2B4, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Avenue West, Montreal, QC H3A 1A1, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - John Cs Breitner
- Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Avenue West, Montreal, QC H3A 1A1, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Gabriel A Devenyi
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Research Centre, 6875 Bd LaSalle CIC Building, Verdun, QC H4H 1R3, Canada; Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Avenue West, Montreal, QC H3A 1A1, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - Christine L Tardif
- Department of Biomedical Engineering, McGill University, Duff Medical Building, 3775 Rue University Suite 316, Montreal, QC H3A 2B4, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, 3801 University St, Montreal, QC H3A2B4, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| | - M Mallar Chakravarty
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Research Centre, 6875 Bd LaSalle CIC Building, Verdun, QC H4H 1R3, Canada; Douglas Mental Health University Institute, 6875 Bd LaSalle, Montreal, QC H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Room 302, Irving Ludmer Building, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada; Department of Biomedical Engineering, McGill University, Duff Medical Building, 3775 Rue University Suite 316, Montreal, QC H3A 2B4, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Avenue West, Montreal, QC H3A 1A1, Canada; Department of Neurology and Neurosurgery, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
2
|
Li JQ, Ma XH, Dai H, Wang CC, Zhang J, Meng XL. Isoliensinine ameliorates cognitive dysfunction in AlCl 3/D-gal-induced Alzheimer's disease-like mice by inhibiting the calcium signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119567. [PMID: 40020795 DOI: 10.1016/j.jep.2025.119567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The embryos of lotus (Nelumbo nucifera Gaertn.) is a famous traditional Chinese medicine used to treat insomnia, memory decline, and dementia for a long time. However, the underlying material basis and mechanisms of this medicine are still unclear. Isoliensinine (IL) is a major alkaloid derived from lotus embryos. Our previous research has demonstrated that IL can exert strong anti-inflammatory and neuroprotective effects in vitro. AIM OF THE STUDY To reveal the underlying therapeutic effect and mechanism of IL on Alzheimer's disease (AD)-like mice induced by AlCl3 and D-galactose (D-gal) in vivo. MATERIALS AND METHODS The AD-like mice were modeled by intragastric injection (i.g.) of AlCl3 (20 mg/kg/day) and intraperitoneal injection (i.p.) of D-gal (120 mg/kg/day) for 8 weeks. Starting from the third week, AD-like mice were treated with IL (1, 3, or 10 mg/kg/day; i.p.) for 6 weeks. Cognitive impairment in AD-like mice was evaluated through some behavioral experiments including nest building, open field, novel object recognition, Y maze, and Morris water maze tests. The cortex and hippocampus (DG, CA1, and CA3) regions were analyzed as follows: Neuronal pathological changes and neurofibrillary tangles (NFTs) formation were observed by hematoxylin-eosin (HE) and silver staining, respectively; The production of Aβ plaques and the activation of microglia and astrocytes were detected by immunohistochemistry; The levels of Ca2+ levels were determined by the ortho-cresolphtalein complexone method. The levels of inflammatory cytokines (TNF-α, IL-6, and IL-1β) were analyzed using the ELISA kits. The expression of CaM, p-CaMKII, Calpain, CDK5, p35/p25, p-Tau, ADAM10, BACE1, PSEN1, APP, Aβ1-42, p-IκBα, and IκBα were evaluated by western blotting. RESULTS IL (1, 3, and 10 mg/kg) treatment effectively ameliorated cognitive impairment in AD-like model mice. IL inhibited the decrease of brain index and body weight in AD-like mice and alleviated neuronal damage in the cortex and hippocampus (DG, CA1, and CA3). IL decreased the levels of Ca2+ and reduce high expression of CaM and Calpain in the cortex and hippocampus of AD-like mice. IL treatment did not affect the expression of CDK5 but inhibited the expression of p-CaMKII and p25/p35, and reduced Tau phosphorylation and NFTs formation. IL also down-regulated the high expression of Aβ1-42 and APP and regulated the expression of APP-cleavage secretase (reducing the expression of BACE1 and PSEN1, while increasing the expression of ADAM10), thereby inhibited the production of Aβ plaques in AD-like mouse brain. Moreover, IL inhibited the phosphorylation and degradation of IκBα, as well as the production of inflammatory cytokines (TNF-α, IL-6, and IL-1β), and prevented the activation of microglia and astrocytes in AD-like mice. CONCLUSIONS IL has a significant therapeutic effect on pathological alterations and cognitive impairment in AlCl3 and D-gal-induced AD-like mice, indicating that IL may have the potential to treat AD. The anti-AD activity of IL may be associated with its regulation of the Ca2+ homeostasis and downstream signaling molecules such as CaM and Calpain.
Collapse
Affiliation(s)
- Jin-Qiu Li
- School of Pharmaceutical Science, Liaoning University, Shenyang, People's Republic of China
| | - Xiao-Han Ma
- School of Pharmaceutical Science, Liaoning University, Shenyang, People's Republic of China
| | - Hui Dai
- School of Pharmaceutical Science, Liaoning University, Shenyang, People's Republic of China
| | - Cheng-Cheng Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang, People's Republic of China
| | - Jing Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang, People's Republic of China
| | - Xue-Lian Meng
- School of Pharmaceutical Science, Liaoning University, Shenyang, People's Republic of China.
| |
Collapse
|
3
|
Xie Z, Situ Y, Deng L, Liang M, Ding H, Guo Z, Xu Q, Liang Z, Shao Z. Identification of therapeutic targets for Alzheimer's Disease Treatment using bioinformatics and machine learning. Sci Rep 2025; 15:3888. [PMID: 39890844 PMCID: PMC11785788 DOI: 10.1038/s41598-025-88134-w] [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: 11/14/2024] [Accepted: 01/24/2025] [Indexed: 02/03/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder that currently lacks effective treatment options. This study aimed to identify potential therapeutic targets for the treatment of AD using comprehensive bioinformatics methods and machine learning algorithms. By integrating differential gene expression analysis, weighted gene co-expression network analysis, Mfuzz clustering, single-cell RNA sequencing, and machine learning algorithms including LASSO regression, SVM-RFE, and random forest, five hub genes related to AD, including PLCB1, NDUFAB1, KRAS, ATP2A2, and CALM3 were identified. PLCB1, in particular, exhibited the highest diagnostic value in AD and showed significant correlation with Braak stages and neuronal expression. Furthermore, Noscapine, PX-316, and TAK-901 were selected as potential therapeutic drugs for AD based on PLCB1. This research provides a comprehensive and reliable method for the discovery of AD therapeutic targets and the construction of diagnostic models, offering important insights and directions for future AD treatment strategies and drug development.
Collapse
Affiliation(s)
- ZhanQiang Xie
- Department of Thoracic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - YongLi Situ
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Li Deng
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Meng Liang
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Hang Ding
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Zhen Guo
- Laboratory of Pathogenic Biology, Guangdong Medical University, Zhanjiang, 524023, China
| | - QinYing Xu
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Zhu Liang
- Department of Thoracic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Zheng Shao
- Department of Parasitology, Guangdong Medical University, Zhanjiang, 524023, China.
| |
Collapse
|
4
|
Lim D, Matute C, Cavaliere F, Verkhratsky A. Neuroglia in neurodegeneration: Alzheimer, Parkinson, and Huntington disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:9-44. [PMID: 40148060 DOI: 10.1016/b978-0-443-19102-2.00012-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The conspicuous rise of chronic neurodegenerative diseases, including Alzheimer (AD), Parkinson (PD), and Huntington (HD) diseases, is currently without disease-modifying therapies and accompanied by an excessive rate of unsuccessful clinical trials. This reflects a profound lack of understanding of the pathogenesis of these diseases, indicating that the current paradigms guiding disease modeling and drug development are in need of reconsideration. The role of neuroglia, namely astrocytes, microglial cells, and oligodendrocytes, in the pathogenesis of neurodegenerative diseases emerged during the last decades. This chapter provides the state-of-the-art update on the changes of astrocytes, microglial cells, and oligodendrocytes in AD, PD, and HD. A growing body of evidence suggests that homeostatic and defensive functions of glial cells are compromised at different disease stages, leading to increased susceptibility of neurons to noxious stimuli, eventually resulting in their malfunction and degeneration. Investments are needed in the generation of novel preclinical models suitable for studying glial pathology, in "humanizing" research, and in-depth investigation of glial cell alterations to slow down and, possibly, halt and prevent the rise of neurodegenerative disease. Targeting glial cells opens new therapeutic avenues to treat AD, PD, and HD.
Collapse
Affiliation(s)
- Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy.
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain
| | - Fabio Cavaliere
- The Basque Biomodels Platform for Human Research (BBioH), Achucarro Basque Center for Neuroscience & Fundación Biofisica Bizkaia, Leioa, Spain
| | - Alexei Verkhratsky
- Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
5
|
Shiga Y, Rangel Olguin AG, El Hajji S, Belforte N, Quintero H, Dotigny F, Alarcon-Martinez L, Krishnaswamy A, Di Polo A. Endoplasmic reticulum stress-related deficits in calcium clearance promote neuronal dysfunction that is prevented by SERCA2 gene augmentation. Cell Rep Med 2024; 5:101839. [PMID: 39615485 PMCID: PMC11722116 DOI: 10.1016/j.xcrm.2024.101839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/25/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024]
Abstract
Disruption of calcium (Ca2+) homeostasis in neurons is a hallmark of neurodegenerative diseases. Here, we investigate the mechanisms leading to Ca2+ dysregulation and ask whether altered Ca2+ dynamics impinge on neuronal stress and circuit dysfunction. Using two-photon microscopy, we show that ocular hypertension, a major risk factor in glaucoma, and optic nerve crush injury disrupt the capacity of retinal neurons to clear cytosolic Ca2+ leading to impaired light-evoked responses. Gene- and protein expression analysis reveal the loss of the sarco-endoplasmic reticulum (ER) Ca2+-ATPase2 pump (SERCA2/ATP2A2) in injured retinal neurons from mice and patients with primary open-angle glaucoma. Pharmacological activation or neuron-specific gene delivery of SERCA2 is sufficient to rescue single-cell Ca2+ dynamics and promote robust survival of damaged neurons. Furthermore, SERCA2 gene supplementation reduces ER stress, reestablishes circuit balance, and restores visual behaviors. Our findings reveal that enhancing the Ca2+ clearance capacity of vulnerable neurons alleviates organelle stress and promotes neurorecovery.
Collapse
Affiliation(s)
- Yukihiro Shiga
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | | | - Sana El Hajji
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Nicolas Belforte
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Heberto Quintero
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Florence Dotigny
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Luis Alarcon-Martinez
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Arjun Krishnaswamy
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, PO box 6128, Station Centre-ville, Montreal, Quebec H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, Quebec H2X 0A9, Canada.
| |
Collapse
|
6
|
Yang Y, Valencia LA, Lu CH, Nakamoto ML, Tsai CT, Liu C, Yang H, Zhang W, Jahed Z, Lee WR, Santoro F, Liou J, Wu JC, Cui B. Plasma membrane curvature regulates the formation of contacts with the endoplasmic reticulum. Nat Cell Biol 2024; 26:1878-1891. [PMID: 39289582 PMCID: PMC11567891 DOI: 10.1038/s41556-024-01511-x] [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: 11/20/2023] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
Contact sites between the endoplasmic reticulum (ER) and plasma membrane (PM) play a crucial role in governing calcium regulation and lipid homeostasis. Despite their significance, the factors regulating their spatial distribution on the PM remain elusive. Inspired by observations in cardiomyocytes, where ER-PM contact sites concentrate on tubular PM invaginations known as transverse tubules, we hypothesize that PM curvature plays a role in ER-PM contact formation. Through precise control of PM invaginations, we show that PM curvatures locally induce the formation of ER-PM contacts in cardiomyocytes. Intriguingly, the junctophilin family of ER-PM tethering proteins, specifically expressed in excitable cells, is the key player in this process, whereas the ubiquitously expressed extended synaptotagmin-2 does not show a preference for PM curvature. At the mechanistic level, we find that the low-complexity region (LCR) and membrane occupation and recognition nexus (MORN) motifs of junctophilins can bind independently to the PM, but both the LCR and MORN motifs are required for targeting PM curvatures. By examining the junctophilin interactome, we identify a family of curvature-sensing proteins-Eps15 homology domain-containing proteins-that interact with the MORN_LCR motifs and facilitate the preferential tethering of junctophilins to curved PM. These findings highlight the pivotal role of PM curvature in the formation of ER-PM contacts in cardiomyocytes and unveil a mechanism for the spatial regulation of ER-PM contacts through PM curvature modulation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Luis A Valencia
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Chih-Hao Lu
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Melissa L Nakamoto
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Departments of Physiology and Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Wei Zhang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Zeinab Jahed
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Chemical and Nano Engineering, University of California, San Diego, San Diego, CA, USA
| | - Wan-Ru Lee
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, Naples, Italy
- Faculty of Electrical Engineering and Information Technology, RWTH Aachen University, Aachen, Germany
- Institute of Biological Information Processing-Bioelectronics (IBI-3), Forschungszentrum, Jülich, Germany
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
7
|
Liu JY, Yin X, Dong YT. Exploration of the shared gene signatures and molecular mechanisms between Alzheimer's disease and intracranial aneurysm. Sci Rep 2024; 14:24628. [PMID: 39427050 PMCID: PMC11490550 DOI: 10.1038/s41598-024-75694-6] [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: 05/27/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
Although Alzheimer's disease (AD) and intracranial aneurysm (IA) were two different types of diseases that occurred in the brain, ruptured IA (RIA) survivors may experience varying degrees of cognitive dysfunction. Neither AD nor IA is easily recognizable by an early onset so that the incidence of adverse clinical outcomes would be on the rise. Therefore, we focused on the exploration of the shared genes and molecular mechanisms between AD and IA, which would be significant for the efficiency of co-screening and co-diagnosis. Two GEO datasets were selected for the weighted gene co-expression network analysis (WGCNA) and differentially expressed gene screening, obtaining 78 overlapped genes. Next, 9 hub genes were identified by the protein-protein interaction network, including PIK3CA, GAB1, IGF1R, PLCB1, PGR, PDGFRB, PLCE1, FGFR3, and SYNJ1. The interactions among the hub genes, miRNA, and TFs were also explored. Meanwhile, we performed GO and KEGG pathway enrichment analyses for the results of WGCNA and hub genes, which showed that the Ras signaling and Rap1 signaling were the main shared pathogenesis. In conclusion, the present bioinformatics analysis revealed that AD and IA had the shared genes and molecular mechanisms, and these outcomes were associated with inflammation and calcium homeostasis, which could provide research clues for further studies.
Collapse
Affiliation(s)
- Ji-Yun Liu
- Department of Clinical Laboratory, Guiyang Second People's Hospital, Guiyang, People's Republic of China
| | - Xuan Yin
- Department of Women Healthcare, Guiyang Maternal and Child Health Hospital, Guiyang, People's Republic of China
| | - Yang-Ting Dong
- Key Laboratory of Endemic and Ethnic Diseases (Guizhou Medical University) of the Ministry of Education and Provincial Key Laboratory of Medical Molecular Biology, No. 9, Beijing Road, Guiyang, 550004, People's Republic of China.
| |
Collapse
|
8
|
Vorobev S, Yanishevskiy S, Efimtsev S, Sokolov A, Dyachuk V. Development of capillary dysfunction in Alzheimer's disease. Front Aging Neurosci 2024; 16:1458455. [PMID: 39267722 PMCID: PMC11391207 DOI: 10.3389/fnagi.2024.1458455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Alzheimer's disease (AD) is currently considered the major cause of cognitive impairment in older adults. This explains the close attention to the issue of AD research. The pathomorphological basis of the disease is a neurodegenerative process, the early stages of which are formed in the hippocampus and the morphofunctionally deep parts of the temporal lobes of the brain closely related to it. Several hypotheses have been advanced concerning the causes of neurodegeneration: the amyloid hypothesis, the calcium homeostasis impairment hypothesis, the inflammatory hypothesis, and the prion hypothesis. However, these hypotheses cannot explain the early stages of the pathogenesis of neurodegenerative diseases, in particular Alzheimer's disease. This health problem requires further comprehensive study of available data, as well as additional investigations to determine the nature of such a process. In this review, the data on microcirculatory disorders in the capillaries of the hippocampus and mediobasal structures of the temporal lobes of the brain, which may be an initiating factor that triggers neurodegenerative events, are analyzed.
Collapse
Affiliation(s)
- S Vorobev
- Laboratory of Neurogenesis and Neurodevelopmental Disorders, World-Class Research Center for Personalized Medicine, Almazov Center, Saint-Petersburg, Russia
| | - S Yanishevskiy
- Laboratory of Neurogenesis and Neurodevelopmental Disorders, World-Class Research Center for Personalized Medicine, Almazov Center, Saint-Petersburg, Russia
| | - S Efimtsev
- Laboratory of Neurogenesis and Neurodevelopmental Disorders, World-Class Research Center for Personalized Medicine, Almazov Center, Saint-Petersburg, Russia
| | - A Sokolov
- Laboratory of Neurogenesis and Neurodevelopmental Disorders, World-Class Research Center for Personalized Medicine, Almazov Center, Saint-Petersburg, Russia
| | - V Dyachuk
- Laboratory of Neurogenesis and Neurodevelopmental Disorders, World-Class Research Center for Personalized Medicine, Almazov Center, Saint-Petersburg, Russia
| |
Collapse
|
9
|
Bagheri S, Saboury AA, Saso L. Sequence of Molecular Events in the Development of Alzheimer's Disease: Cascade Interactions from Beta-Amyloid to Other Involved Proteins. Cells 2024; 13:1293. [PMID: 39120323 PMCID: PMC11312137 DOI: 10.3390/cells13151293] [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: 06/11/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Alzheimer's disease is the primary neurodegenerative disease affecting the elderly population. Despite the first description of its pathology over a century ago, its precise cause and molecular mechanism remain unknown. Numerous factors, including beta-amyloid, tau protein, the APOEε4 gene, and different metals, have been extensively investigated in relation to this disease. However, none of them have been proven to have a decisive causal relationship. Furthermore, no single theory has successfully integrated these puzzle pieces thus far. In this review article, we propose the most probable molecular mechanism for AD, which clearly shows the relationship between the main aspects of the disease, and addresses fundamental questions such as: Why is aging the major risk factor for the disease? Are amyloid plaques and tau tangles the causes or consequences of AD? Why are the distributions of senile plaques and tau tangles in the brain different and independent of each other? Why is the APOEε4 gene a risk factor for AD? Finally, why is the disease more prevalent in women?
Collapse
Affiliation(s)
- Soghra Bagheri
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, 00185 Rome, Italy;
| |
Collapse
|
10
|
Wang L, Zhang J, Li W, Zhang X, Yokoyama T, Sakamoto M, Wang Y. The A-kinase anchoring protein Yotiao decrease the ER calcium content by inhibiting the store operated calcium entry. Cell Calcium 2024; 121:102906. [PMID: 38781694 DOI: 10.1016/j.ceca.2024.102906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
The meticulous regulation of ER calcium (Ca2+) homeostasis is indispensable for the proper functioning of numerous cellular processes. Disrupted ER Ca2+ balance is implicated in diverse diseases, underscoring the need for a systematic exploration of its regulatory factors in cells. Our recent genomic-scale screen identified a scaffolding protein A-kinase anchoring protein 9 (AKAP9) as a regulator of ER Ca2+ levels, but the underlying molecular mechanisms remain elusive. Here, we reveal that Yotiao, the smallest splicing variant of AKAP9 decreased ER Ca2+ content in animal cells. Additional testing using a combination of Yotiao truncations, knock-out cells and pharmacological tools revealed that, Yotiao does not require most of its interactors, including type 1 inositol 1,4,5-trisphosphate receptors (IP3R1), protein kinase A (PKA), protein phosphatase 1 (PP1), adenylyl cyclase type 2 (AC2) and so on, to reduce ER Ca2+ levels. However, adenylyl cyclase type 9 (AC9), which is known to increases its cAMP generation upon interaction with Yotiao for the modulation of potassium channels, plays an essential role for Yotiao's ER-Ca2+-lowering effect. Mechanistically, Yotiao may work through AC9 to act on Orai1-C terminus and suppress store operated Ca2+ entry, resulting in reduced ER Ca2+ levels. These findings not only enhance our comprehension of the interplay between Yotiao and AC9 but also contribute to a more intricate understanding of the finely tuned mechanisms governing ER Ca2+ homeostasis.
Collapse
Affiliation(s)
- Liuqing Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jiaxuan Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Wanjie Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Xiaoyan Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Tatsushi Yokoyama
- Department of Optical Neural and Molecular Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Masayuki Sakamoto
- Department of Optical Neural and Molecular Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China.
| |
Collapse
|
11
|
Yang Y, Valencia LA, Lu CH, Nakamoto ML, Tsai CT, Liu C, Yang H, Zhang W, Jahed Z, Lee WR, Santoro F, Liou J, Wu JC, Cui B. Membrane Curvature Promotes ER-PM Contact Formation via Junctophilin-EHD Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.29.601287. [PMID: 38979311 PMCID: PMC11230412 DOI: 10.1101/2024.06.29.601287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Contact sites between the endoplasmic reticulum (ER) and the plasma membrane (PM) play a crucial role in governing calcium regulation and lipid homeostasis. Despite their significance, the factors regulating their spatial distribution on the PM remain elusive. Inspired by observations in cardiomyocytes, where ER-PM contact sites concentrate on tubular PM invaginations known as transverse tubules (T-tubules), we hypothesize that the PM curvature plays a role in ER-PM contact formation. Through precise control of PM invaginations, we show that PM curvatures locally induce the formation of ER-PM contacts in cardiomyocytes. Intriguingly, the junctophilin family of ER-PM tethering proteins, specifically expressed in excitable cells, is the key player in this process, while the ubiquitously expressed extended synaptotagmin 2 does not show a preference for PM curvature. At the mechanistic level, we find that the low complexity region (LCR) and the MORN motifs of junctophilins can independently bind to the PM, but both the LCR and MORN motifs are required for targeting PM curvatures. By examining the junctophilin interactome, we identify a family of curvature-sensing proteins, Eps15-homology domain containing proteins (EHDs), that interact with the MORN_LCR motifs and facilitate junctophilins' preferential tethering to curved PM. These findings highlight the pivotal role of PM curvature in the formation of ER-PM contacts in cardiomyocytes and unveil a novel mechanism for the spatial regulation of ER-PM contacts through PM curvature modulation.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Luis A. Valencia
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Chih-Hao Lu
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Melissa L. Nakamoto
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Present address: Department of Physiology and Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Present address: Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Wei Zhang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| | - Zeinab Jahed
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Present address: Department of Nanoengineering, Jacobs School of Engineering, University of California, San Diego, CA, USA
| | - Wan-Ru Lee
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Faculty of Electrical Engineering and IT, RWTH, Aachen 52074, Germany
- Institute of Biological Information Processing—Bioelectronics, IBI-3, Forschungszentrum, Juelich 52428, Germany
| | - Jen Liou
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Medicine (Division of Cardiology), Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University; Stanford, CA, USA
| |
Collapse
|
12
|
Shi Q, Sun X, Zhang H, Yang L, Fu Y, Wang G, Su Y, Li W, Li W. PLC-CN-NFAT1 signaling-mediated Aβ and IL-1β crosstalk synergistically promotes hippocampal neuronal damage. Int Immunopharmacol 2024; 134:112259. [PMID: 38749336 DOI: 10.1016/j.intimp.2024.112259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/18/2024] [Accepted: 05/10/2024] [Indexed: 06/03/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. Neuronal calcium overload plays an important role in Aβ deposition and neuroinflammation, which are strongly associated with AD. However, the specific mechanisms by which calcium overload contributes to neuroinflammation and AD and the relationship between them have not been elucidated. Phospholipase C (PLC) is involved in regulation of calcium homeostasis, and CN-NFAT1 signaling is dependent on intracellular Ca2+ ([Ca2+]i) to regulate transcription of genes. Therefore, we hypothesized that the PLC-CN-NFAT1 signaling might mediate the interaction between Aβ and inflammation to promote neuronal injury in AD. In this experiment, the results showed that the levels of Aβ, IL-1β and [Ca2+]i in the hippocampal primary neurons of APP/PS1 mice (APP neurons) were significantly increased. IL-1β exposure also significantly increased Aβ and [Ca2+]i in HT22 cells, suggesting a close association between Aβ and IL-1β in the development of AD. Furthermore, PLC activation induced significant calcium homeostasis imbalance, cell apoptosis, Aβ and ROS production, and significantly increased expressions of CN and NFAT1, while PLC inhibitor significantly reversed these changes in APP neurons and IL-1β-induced HT22 cells. Further results indicated that PLC activation significantly increased the expressions of NOX2, APP, BACE1, and NCSTN, which were inhibited by PLC inhibitor in APP neurons and IL-1β-induced HT22 cells. All indications point to a synergistic interaction between Aβ and IL-1β by activating the PLC-CN-NFAT1 signal, ultimately causing a vicious cycle, resulting in neuronal damage in AD. The study may provide a new idea and target for treatment of AD.
Collapse
Affiliation(s)
- Qifeng Shi
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Xiangyu Sun
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Hui Zhang
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Liu Yang
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yinglin Fu
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Guohang Wang
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yong Su
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei 230032, Anhui, China
| | - Weiping Li
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
13
|
Yasamineh S, Mehrabani FJ, Derafsh E, Danihiel Cosimi R, Forood AMK, Soltani S, Hadi M, Gholizadeh O. Potential Use of the Cholesterol Transfer Inhibitor U18666A as a Potent Research Tool for the Study of Cholesterol Mechanisms in Neurodegenerative Disorders. Mol Neurobiol 2024; 61:3503-3527. [PMID: 37995080 DOI: 10.1007/s12035-023-03798-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/03/2023] [Indexed: 11/24/2023]
Abstract
Cholesterol is an essential component of mammalian cell membranes and a precursor for crucial signaling molecules. The brain contains the highest level of cholesterol in the body, and abnormal cholesterol metabolism links to many neurodegenerative disorders. The results indicate that faulty cholesterol metabolism is a common feature among people living with neurodegenerative conditions. The researchers suggest that restoring cholesterol levels may become a beneficial new strategy in treating certain neurodegenerative conditions. Several neurodegenerative disorders, such as Alzheimer's disease (AD), Niemann-Pick type C (NPC) disease, and Parkinson's disease (PD), have been connected to abnormalities in brain cholesterol metabolism. Consequently, using a lipid research tool is vital to study further and understand the effect of lipids in neurodegenerative disorders such as NPC, AD, PD, and Huntington's disease (HD). U18666A, also known as 3-(2-(diethylamino) ethoxy) androst-5-en-17-one, is a pharmaceutical drug that suppresses cholesterol trafficking and is a well-known class-2 amphiphile. U18666A has performed many functions, allowing for essential discoveries in lipid studies and shedding light on the pathophysiology of neurodegenerative disorders. Additionally, U18666A prevented the downregulation of low-density lipoprotein (LDL) receptors that are induced by LDL and led to the buildup of cholesterol in lysosomes. Numerous studies show that U18666A impacts the function of cholesterol trafficking to control the metabolism and transport of amyloid precursor proteins (APPs). Treating cortical neurons with U18666A may provide a new in vitro model system for studying the underlying molecular process of NPC, AD, HD, and PD. In this article, we review the mechanism and function of U18666A as a vital tool for studying cholesterol mechanisms in neurological diseases related to abnormal cholesterol metabolism, such as AD, NPC, HD, and PD.
Collapse
Affiliation(s)
| | | | - Ehsan Derafsh
- Windsor University School of Medicine, Cayon, Saint Kitts and Nevis
| | | | | | - Siamak Soltani
- Department of Forensic Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Meead Hadi
- Department Of Microbiology, Faculty of Basic Sciences, Tehran Central Branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
14
|
Meimetis N, Lauffenburger DA, Nilsson A. Inference of drug off-target effects on cellular signaling using interactome-based deep learning. iScience 2024; 27:109509. [PMID: 38591003 PMCID: PMC11000001 DOI: 10.1016/j.isci.2024.109509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/04/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Many diseases emerge from dysregulated cellular signaling, and drugs are often designed to target specific signaling proteins. Off-target effects are, however, common and may ultimately result in failed clinical trials. Here we develop a computer model of the cell's transcriptional response to drugs for improved understanding of their mechanisms of action. The model is based on ensembles of artificial neural networks and simultaneously infers drug-target interactions and their downstream effects on intracellular signaling. With this, it predicts transcription factors' activities, while recovering known drug-target interactions and inferring many new ones, which we validate with an independent dataset. As a case study, we analyze the effects of the drug Lestaurtinib on downstream signaling. Alongside its intended target, FLT3, the model predicts an inhibition of CDK2 that enhances the downregulation of the cell cycle-critical transcription factor FOXM1. Our approach can therefore enhance our understanding of drug signaling for therapeutic design.
Collapse
Affiliation(s)
- Nikolaos Meimetis
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Avlant Nilsson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Cell and Molecular Biology, SciLifeLab, Karolinska Institutet, Stockholm, Sweden
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, SE 41296, Sweden
| |
Collapse
|
15
|
Zhang Q, Liu J, Liu H, Ao L, Xi Y, Chen D. Genome-wide epistasis analysis reveals gene-gene interaction network on an intermediate endophenotype P-tau/Aβ 42 ratio in ADNI cohort. Sci Rep 2024; 14:3984. [PMID: 38368488 PMCID: PMC10874417 DOI: 10.1038/s41598-024-54541-8] [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: 10/22/2023] [Accepted: 02/14/2024] [Indexed: 02/19/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia in the elderly worldwide. The exact etiology of AD, particularly its genetic mechanisms, remains incompletely understood. Traditional genome-wide association studies (GWAS), which primarily focus on single-nucleotide polymorphisms (SNPs) with main effects, provide limited explanations for the "missing heritability" of AD, while there is growing evidence supporting the important role of epistasis. In this study, we performed a genome-wide SNP-SNP interaction detection using a linear regression model and employed multiple GPUs for parallel computing, significantly enhancing the speed of whole-genome analysis. The cerebrospinal fluid (CSF) phosphorylated tau (P-tau)/amyloid-[Formula: see text] (A[Formula: see text]) ratio was used as a quantitative trait (QT) to enhance statistical power. Age, gender, and clinical diagnosis were included as covariates to control for potential non-genetic factors influencing AD. We identified 961 pairs of statistically significant SNP-SNP interactions, explaining a high-level variance of P-tau/A[Formula: see text] level, all of which exhibited marginal main effects. Additionally, we replicated 432 previously reported AD-related genes and found 11 gene-gene interaction pairs overlapping with the protein-protein interaction (PPI) network. Our findings may contribute to partially explain the "missing heritability" of AD. The identified subnetwork may be associated with synaptic dysfunction, Wnt signaling pathway, oligodendrocytes, inflammation, hippocampus, and neuronal cells.
Collapse
Affiliation(s)
- Qiushi Zhang
- School of Computer Science, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China
| | - Junfeng Liu
- School of Computer Science, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China
| | - Hongwei Liu
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, 145 Nantong Street, Harbin, China
| | - Lang Ao
- School of Computer Science, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China
| | - Yang Xi
- School of Computer Science, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China
| | - Dandan Chen
- School of Automation Engineering, Northeast Electric Power University, 169 Changchun Street, Jilin, 132012, China.
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, 145 Nantong Street, Harbin, China.
| |
Collapse
|
16
|
Sneyers F, Speelman-Rooms F, Verhelst SHL, Bootman MD, Bultynck G. Cellular effects of BAPTA: Are they only about Ca 2+ chelation? BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119589. [PMID: 37739271 DOI: 10.1016/j.bbamcr.2023.119589] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 09/24/2023]
Abstract
Intracellular Ca2+ signals play a vital role in a broad range of cell biological and physiological processes in all eukaryotic cell types. Dysregulation of Ca2+ signaling has been implicated in numerous human diseases. Over the past four decades, the understanding of how cells use Ca2+ as a messenger has flourished, largely because of the development of reporters that enable visualization of Ca2+ signals in different cellular compartments, and tools that can modulate cellular Ca2+ signaling. One such tool that is frequently used is BAPTA; a fast, high-affinity Ca2+-chelating molecule. By making use of a cell-permeable acetoxymethyl ester (AM) variant, BAPTA can be readily loaded into the cytosol of cells (referred to as BAPTAi), where it is trapped and able to buffer changes in cytosolic Ca2+. Due to the ease of loading of the AM version of BAPTA, this reagent has been used in hundreds of studies to probe the role of Ca2+ signaling in specific processes. As such, for decades, researchers have almost universally attributed changes in biological processes caused by BAPTAi to the involvement of Ca2+ signaling. However, BAPTAi has often been used without any form of control, and in many cases has neither been shown to be retained in cells for the duration of experiments nor to buffer any Ca2+ signals. Moreover, increasing evidence points to off-target cellular effects of BAPTA that are clearly not related to Ca2+ chelation. Here, we briefly introduce Ca2+ signaling and the history of Ca2+ chelators and fluorescent Ca2+ indicators. We highlight Ca2+-independent effects of BAPTAi on a broad range of molecular targets and describe some of BAPTAi's impacts on cell functions that occur independently of its Ca2+-chelating properties. Finally, we propose strategies for determining whether Ca2+ chelation, the binding of other metal ions, or off-target interactions with cell components are responsible for BAPTAi's effect on a particular process and suggest some future research directions.
Collapse
Affiliation(s)
- Flore Sneyers
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, B-3000 Leuven, Belgium
| | - Femke Speelman-Rooms
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, B-3000 Leuven, Belgium; KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, Herestraat 49 box 901b, B-3000 Leuven, Belgium
| | - Steven H L Verhelst
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, Herestraat 49 box 901b, B-3000 Leuven, Belgium
| | - Martin D Bootman
- The Open University, Cancer Research Group, School of Life, Health and Chemical Sciences, Milton Keynes, UK
| | - Geert Bultynck
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
17
|
Parkkinen I, Their A, Asghar MY, Sree S, Jokitalo E, Airavaara M. Pharmacological Regulation of Endoplasmic Reticulum Structure and Calcium Dynamics: Importance for Neurodegenerative Diseases. Pharmacol Rev 2023; 75:959-978. [PMID: 37127349 DOI: 10.1124/pharmrev.122.000701] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 05/03/2023] Open
Abstract
The endoplasmic reticulum (ER) is the largest organelle of the cell, composed of a continuous network of sheets and tubules, and is involved in protein, calcium (Ca2+), and lipid homeostasis. In neurons, the ER extends throughout the cell, both somal and axodendritic compartments, and is highly important for neuronal functions. A third of the proteome of a cell, secreted and membrane-bound proteins, are processed within the ER lumen and most of these proteins are vital for neuronal activity. The brain itself is high in lipid content, and many structural lipids are produced, in part, by the ER. Cholesterol and steroid synthesis are strictly regulated in the ER of the blood-brain barrier protected brain cells. The high Ca2+ level in the ER lumen and low cytosolic concentration is needed for Ca2+-based intracellular signaling, for synaptic signaling and Ca2+ waves, and for preparing proteins for correct folding in the presence of high Ca2+ concentrations to cope with the high concentrations of extracellular milieu. Particularly, ER Ca2+ is controlled in axodendritic areas for proper neurito- and synaptogenesis and synaptic plasticity and remodeling. In this review, we cover the physiologic functions of the neuronal ER and discuss it in context of common neurodegenerative diseases, focusing on pharmacological regulation of ER Ca2+ Furthermore, we postulate that heterogeneity of the ER, its protein folding capacity, and ensuring Ca2+ regulation are crucial factors for the aging and selective vulnerability of neurons in various neurodegenerative diseases. SIGNIFICANCE STATEMENT: Endoplasmic reticulum (ER) Ca2+ regulators are promising therapeutic targets for degenerative diseases for which efficacious drug therapies do not exist. The use of pharmacological probes targeting maintenance and restoration of ER Ca2+ can provide restoration of protein homeostasis (e.g., folding of complex plasma membrane signaling receptors) and slow down the degeneration process of neurons.
Collapse
Affiliation(s)
- Ilmari Parkkinen
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Anna Their
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Muhammad Yasir Asghar
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Sreesha Sree
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Eija Jokitalo
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| |
Collapse
|
18
|
Vatsal VH, Jha BK, Singh TP. To study the effect of ER flux with buffer on the neuronal calcium. EUROPEAN PHYSICAL JOURNAL PLUS 2023; 138:494. [PMID: 37304245 PMCID: PMC10240135 DOI: 10.1140/epjp/s13360-023-04077-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/07/2023] [Indexed: 06/13/2023]
Abstract
Calcium signaling is decisive for cellular functions. This calcium random walk stipulates neuronal functions. Calcium concentration could provoke gene transcription, apoptosis, neuronal plasticity, etc. A malformation in calcium could change the neuron's intracellular behavior. Calcium concentration balancing is a complex cellular mechanism. This occurrence can be handled with the Caputo fractional reaction-diffusion equation. In this mathematical modeling, we have included the STIM-Orai mechanism and Endoplasmic Reticulum (ER) flux, Inositol Triphosphate Receptor (IPR), SERCA, plasma membrane flux, voltage-gated calcium entry, and different buffer interactions. A hybrid integral transform and Green's function approach were taken to solve the initial boundary problem. A closed-form solution of a Mittag-Leffler family function plotted using MATLAB software. Different parameters impact changes in the spatiotemporal behavior of the calcium concentration. Specific roles of organelles involved in Alzheimer's disease-affected neurons are computed. Ethylene glycol tetraacetic acid (EGTA), 1,2-bis(o-aminophenoxy)ethane N,N,N,N-tetraacetic acid (BAPTA), and S100B protein effects are also observed. In all simulations, we can say S100B and the STIM-Orai effect cannot be neglected. This model lights up the different approaches for calcium signaling pathway simulation. As a consequence, we determine that a generalized reaction-diffusion approach is a better fit realistic model.
Collapse
Affiliation(s)
- Vora Hardagna Vatsal
- Department of Mathematics, Pandit Deendayal Energy University, Gandhinagar, 382007 Gujarat India
| | - Brajesh Kumar Jha
- Department of Mathematics, Pandit Deendayal Energy University, Gandhinagar, 382007 Gujarat India
| | - Tajinder Pal Singh
- Department of Mathematics, Pandit Deendayal Energy University, Gandhinagar, 382007 Gujarat India
| |
Collapse
|
19
|
Baracaldo-Santamaría D, Avendaño-Lopez SS, Ariza-Salamanca DF, Rodriguez-Giraldo M, Calderon-Ospina CA, González-Reyes RE, Nava-Mesa MO. Role of Calcium Modulation in the Pathophysiology and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24109067. [PMID: 37240413 DOI: 10.3390/ijms24109067] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease and the most frequent cause of progressive dementia in senior adults. It is characterized by memory loss and cognitive impairment secondary to cholinergic dysfunction and N-methyl-D-aspartate (NMDA)-mediated neurotoxicity. Intracellular neurofibrillary tangles, extracellular plaques composed of amyloid-β (Aβ), and selective neurodegeneration are the anatomopathological hallmarks of this disease. The dysregulation of calcium may be present in all the stages of AD, and it is associated with other pathophysiological mechanisms, such as mitochondrial failure, oxidative stress, and chronic neuroinflammation. Although the cytosolic calcium alterations in AD are not completely elucidated, some calcium-permeable channels, transporters, pumps, and receptors have been shown to be involved at the neuronal and glial levels. In particular, the relationship between glutamatergic NMDA receptor (NMDAR) activity and amyloidosis has been widely documented. Other pathophysiological mechanisms involved in calcium dyshomeostasis include the activation of L-type voltage-dependent calcium channels, transient receptor potential channels, and ryanodine receptors, among many others. This review aims to update the calcium-dysregulation mechanisms in AD and discuss targets and molecules with therapeutic potential based on their modulation.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Sara Sofia Avendaño-Lopez
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Daniel Felipe Ariza-Salamanca
- Medical and Health Sciences Education Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mateo Rodriguez-Giraldo
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos A Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- Grupo de Investigación en Ciencias Biomédicas Aplicadas (UR Biomed), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mauricio O Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
20
|
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
|
21
|
Chen WB, Wang YX, Wang HG, An D, Sun D, Li P, Zhang T, Lu WG, Liu YQ. Role of TPEN in Amyloid-β 25-35-Induced Neuronal Damage Correlating with Recovery of Intracellular Zn 2+ and Intracellular Ca 2+ Overloading. Mol Neurobiol 2023:10.1007/s12035-023-03322-x. [PMID: 37059931 DOI: 10.1007/s12035-023-03322-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/17/2023] [Indexed: 04/16/2023]
Abstract
The overproduction of neurotoxic amyloid-β (Aβ) peptides in the brain is a hallmark of Alzheimer's disease (AD). To determine the role of intracellular zinc ion (iZn2+) dysregulation in mediating Aβ-related neurotoxicity, this study aimed to investigate whether N, N, N', N'‑tetrakis (2‑pyridylmethyl) ethylenediamine (TPEN), a Zn2+‑specific chelator, could attenuate Aβ25-35‑induced neurotoxicity and the underlying mechanism. We used the 3-(4, 5-dimethyl-thiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay to measure the viability of primary hippocampal neurons. We also determined intracellular Zn2+ and Ca2+ concentrations, mitochondrial and lysosomal functions, and intracellular reactive oxygen species (ROS) content in hippocampal neurons using live-cell confocal imaging. We detected L-type voltage-gated calcium channel currents (L-ICa) in hippocampal neurons using the whole‑cell patch‑clamp technique. Furthermore, we measured the mRNA expression levels of proteins related to the iZn2+ buffer system (ZnT-3, MT-3) and voltage-gated calcium channels (Cav1.2, Cav1.3) in hippocampal neurons using RT-PCR. The results showed that TPEN attenuated Aβ25-35‑induced neuronal death, relieved the Aβ25-35‑induced increase in intracellular Zn2+ and Ca2+ concentrations; reversed the Aβ25-35‑induced increase in ROS content, the Aβ25-35‑induced increase in the L-ICa peak amplitude at different membrane potentials, the Aβ25-35‑induced the dysfunction of the mitochondria and lysosomes, and the Aβ25-35‑induced decrease in ZnT-3 and MT-3 mRNA expressions; and increased the Cav1.2 mRNA expression in the hippocampal neurons. These results suggest that TPEN, the Zn2+-specific chelator, attenuated Aβ25-35‑induced neuronal damage, correlating with the recovery of intracellular Zn2+ and modulation of abnormal Ca2+-related signaling pathways.
Collapse
Affiliation(s)
- Wen-Bo Chen
- College of Life Sciences, Nankai University, Tianjin, 300071, China
- School of Basic Medical Science, Henan University, Kaifeng, 475004, China
| | - Yu-Xiang Wang
- Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Hebei University of Chinese Medicine, Shijiazhuang, 050200, Hebei, China
| | - Hong-Gang Wang
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Di An
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Dan Sun
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Pan Li
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Department of Neurology, Tianjin Huanhu Hospital Affiliated to Nankai University, Tianjin, China
| | - Tao Zhang
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Wan-Ge Lu
- College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yan-Qiang Liu
- College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
22
|
Lim D, Tapella L, Dematteis G, Genazzani AA, Corazzari M, Verkhratsky A. The endoplasmic reticulum stress and unfolded protein response in Alzheimer's disease: a calcium dyshomeostasis perspective. Ageing Res Rev 2023; 87:101914. [PMID: 36948230 DOI: 10.1016/j.arr.2023.101914] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/03/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
Protein misfolding is prominent in early cellular pathology of Alzheimer's disease (AD), implicating pathophysiological significance of endoplasmic reticulum stress/unfolded protein response (ER stress/UPR) and highlighting it as a target for drug development. Experimental data from animal AD models and observations on human specimens are, however, inconsistent. ER stress and associated UPR are readily observed in in vitro AD cellular models and in some AD model animals. In the human brain, components and markers of ER stress as well as UPR transducers are observed at Braak stages III-VI associated with severe neuropathology and neuronal death. The picture, however, is further complicated by the brain region- and cell type-specificity of the AD-related pathology. Terms 'disturbed' or 'non-canonical' ER stress/UPR were used to describe the discrepancies between experimental data and the classic ER stress/UPR cascade. Here we discuss possible 'disturbing' or 'interfering' factors which may modify ER stress/UPR in the early AD pathogenesis. We focus on the dysregulation of the ER Ca2+ homeostasis, store-operated Ca2+ entry, and the interaction between the ER and mitochondria. We suggest that a detailed study of the CNS cell type-specific alterations of Ca2+ homeostasis in early AD may deepen our understanding of AD-related dysproteostasis.
Collapse
Affiliation(s)
- Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy.
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Via Bovio 6, 28100, Novara, Italy
| | - Marco Corazzari
- Department of Health Science (DSS), Center for Translational Research on Autoimmune and Allergic Disease (CAAD) & Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale "Amedeo Avogadro"
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain & Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania; Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
| |
Collapse
|
23
|
Sreenivasamurthy S, Laul M, Zhao N, Kim T, Zhu D. Current progress of cerebral organoids for modeling Alzheimer's disease origins and mechanisms. Bioeng Transl Med 2023; 8:e10378. [PMID: 36925717 PMCID: PMC10013781 DOI: 10.1002/btm2.10378] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/06/2022] [Accepted: 07/16/2022] [Indexed: 11/06/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disease that has emerged as a leading risk factor for dementia associated with increasing age. Two-dimensional (2D) cell culture and animal models, which have been used to analyze AD pathology and search for effective treatments for decades, have significantly contributed to our understanding of the mechanism of AD. Despite their successes, 2D and animal models can only capture a fraction of AD mechanisms due to their inability to recapitulate human brain-specific tissue structure, function, and cellular diversity. Recently, the emergence of three-dimensional (3D) cerebral organoids using tissue engineering and induced pluripotent stem cell technology has paved the way to develop models that resemble features of human brain tissue more accurately in comparison to prior models. In this review, we focus on summarizing key research strategies for engineering in vitro 3D human brain-specific models, major discoveries from using AD cerebral organoids, and its future perspectives.
Collapse
Affiliation(s)
- Sai Sreenivasamurthy
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew YorkUSA
| | - Mahek Laul
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew YorkUSA
| | - Nan Zhao
- Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Tiffany Kim
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew YorkUSA
| | - Donghui Zhu
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew YorkUSA
| |
Collapse
|
24
|
Joshi M, Joshi S, Khambete M, Degani M. Role of calcium dysregulation in Alzheimer's disease and its therapeutic implications. Chem Biol Drug Des 2023; 101:453-468. [PMID: 36373976 DOI: 10.1111/cbdd.14175] [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: 07/28/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022]
Abstract
The increasing incidence of Alzheimer's disease (AD) coupled with the lack of therapeutics to address the underlying pathology of the disease has necessitated the need for exploring newer targets. Calcium dysregulation represents a relatively newer target associated with AD. Ca+2 serves as an important cellular messenger in neurons. The concentration of the Ca+2 ion needs to be regulated at optimal concentrations intracellularly for normal functioning of the neurons. This is achieved with the help of mitochondria, endoplasmic reticulum, and neuronal plasma membrane channel proteins. Disruption in normal calcium homeostasis can induce formation of amyloid beta plaques, accumulation of neurofibrillary tangles, and dysfunction of synaptic plasticity, which in turn can affect calcium homeostasis further, thus forming a vicious cycle. Hence, understanding calcium dysregulation can prove to be a key to develop newer therapeutics. This review provides detailed account of physiology of calcium homeostasis and its dysregulation associated with AD. Further, with an understanding of various receptors and organelles involved in these pathways, the review also discusses various calcium channel blockers explored in AD hand in hand with some multitarget molecules addressing calcium as one of the targets.
Collapse
Affiliation(s)
- Maithili Joshi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Siddhi Joshi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Mihir Khambete
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Mariam Degani
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| |
Collapse
|
25
|
Walters GC, Usachev YM. Mitochondrial calcium cycling in neuronal function and neurodegeneration. Front Cell Dev Biol 2023; 11:1094356. [PMID: 36760367 PMCID: PMC9902777 DOI: 10.3389/fcell.2023.1094356] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023] Open
Abstract
Mitochondria are essential for proper cellular function through their critical roles in ATP synthesis, reactive oxygen species production, calcium (Ca2+) buffering, and apoptotic signaling. In neurons, Ca2+ buffering is particularly important as it helps to shape Ca2+ signals and to regulate numerous Ca2+-dependent functions including neuronal excitability, synaptic transmission, gene expression, and neuronal toxicity. Over the past decade, identification of the mitochondrial Ca2+ uniporter (MCU) and other molecular components of mitochondrial Ca2+ transport has provided insight into the roles that mitochondrial Ca2+ regulation plays in neuronal function in health and disease. In this review, we discuss the many roles of mitochondrial Ca2+ uptake and release mechanisms in normal neuronal function and highlight new insights into the Ca2+-dependent mechanisms that drive mitochondrial dysfunction in neurologic diseases including epilepsy, Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. We also consider how targeting Ca2+ uptake and release mechanisms could facilitate the development of novel therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Grant C. Walters
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
| | - Yuriy M. Usachev
- Department of Neuroscience and Pharmacology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
26
|
He YJ, Cong L, Liang SL, Ma X, Tian JN, Li H, Wu Y. Discovery and validation of Ferroptosis-related molecular patterns and immune characteristics in Alzheimer's disease. Front Aging Neurosci 2022; 14:1056312. [PMID: 36506471 PMCID: PMC9727409 DOI: 10.3389/fnagi.2022.1056312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022] Open
Abstract
Background To date, the pathogenesis of Alzheimer's disease is still not fully elucidated. Much evidence suggests that Ferroptosis plays a crucial role in the pathogenesis of AD, but little is known about its molecular immunological mechanisms. Therefore, this study aims to comprehensively analyse and explore the molecular mechanisms and immunological features of Ferroptosis-related genes in the pathogenesis of AD. Materials and methods We obtained the brain tissue dataset for AD from the GEO database and downloaded the Ferroptosis-related gene set from FerrDb for analysis. The most relevant Hub genes for AD were obtained using two machine learning algorithms (Least absolute shrinkage and selection operator (LASSO) and multiple support vector machine recursive feature elimination (mSVM-RFE)). The study of the Hub gene was divided into two parts. In the first part, AD patients were genotyped by unsupervised cluster analysis, and the different clusters' immune characteristics were analysed. A PCA approach was used to quantify the FRGscore. In the second part: we elucidate the biological functions involved in the Hub genes and their role in the immune microenvironment by integrating algorithms (GSEA, GSVA and CIBERSORT). Analysis of Hub gene-based drug regulatory networks and mRNA-miRNA-lncRNA regulatory networks using Cytoscape. Hub genes were further analysed using logistic regression models. Results Based on two machine learning algorithms, we obtained a total of 10 Hub genes. Unsupervised clustering successfully identified two different clusters, and immune infiltration analysis showed a significantly higher degree of immune infiltration in type A than in type B, indicating that type A may be at the peak of AD neuroinflammation. Secondly, a Hub gene-based Gene-Drug regulatory network and a ceRNA regulatory network were successfully constructed. Finally, a logistic regression algorithm-based AD diagnosis model and Nomogram diagram were developed. Conclusion Our study provides new insights into the role of Ferroptosis-related molecular patterns and immune mechanisms in AD, as well as providing a theoretical basis for the addition of diagnostic markers for AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yun Wu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
27
|
Intracellular Injection of Brain Extracts from Alzheimer's Disease Patients Triggers Unregulated Ca 2+ Release from Intracellular Stores That Hinders Cellular Bioenergetics. Cells 2022; 11:cells11223630. [PMID: 36429057 PMCID: PMC9688564 DOI: 10.3390/cells11223630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022] Open
Abstract
Strong evidence indicates that amyloid beta (Aβ) inflicts its toxicity in Alzheimer's disease (AD) by promoting uncontrolled elevation of cytosolic Ca2+ in neurons. We have previously shown that synthetic Aβ42 oligomers stimulate abnormal intracellular Ca2+ release from the endoplasmic reticulum stores, suggesting that a similar mechanism of Ca2+ toxicity may be common to the endogenous Aβs oligomers. Here, we use human postmortem brain extracts from AD-affected patients and test their ability to trigger Ca2+ fluxes when injected intracellularly into Xenopus oocytes. Immunological characterization of the samples revealed the elevated content of soluble Aβ oligomers only in samples from AD patients. Intracellular injection of brain extracts from control patients failed to trigger detectable changes in intracellular Ca2+. Conversely, brain extracts from AD patients triggered Ca2+ events consisting of local and global Ca2+ fluorescent transients. Pre-incubation with either the conformation-specific OC antiserum or caffeine completely suppressed the brain extract's ability to trigger cytosolic Ca2+ events. Computational modeling suggests that these Ca2+ fluxes may impair cells bioenergetic by affecting ATP and ROS production. These results support the hypothesis that Aβ oligomers contained in neurons of AD-affected brains may represent the toxic agents responsible for neuronal malfunctioning and death associated with the disruption of Ca2+ homeostasis.
Collapse
|
28
|
Gopalakrishna R, Lin CY, Oh A, Le C, Yang S, Hicks A, Kindy MS, Mack WJ, Bhat NR. cAMP-induced decrease in cell-surface laminin receptor and cellular prion protein attenuates amyloid-β uptake and amyloid-β-induced neuronal cell death. FEBS Lett 2022; 596:2914-2927. [PMID: 35971617 PMCID: PMC9712173 DOI: 10.1002/1873-3468.14467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/21/2022] [Accepted: 07/31/2022] [Indexed: 02/07/2023]
Abstract
Previous studies have shown that amyloid-β oligomers (AβO) bind with high affinity to cellular prion protein (PrPC ). The AβO-PrPC complex binds to cell-surface co-receptors, including the laminin receptor (67LR). Our current studies revealed that in Neuroscreen-1 cells, 67LR is the major co-receptor involved in the cellular uptake of AβO and AβΟ-induced cell death. Both pharmacological (dibutyryl-cAMP, forskolin and rolipram) and physiological (pituitary adenylate cyclase-activating polypeptide) cAMP-elevating agents decreased cell-surface PrPC and 67LR, thereby attenuating the uptake of AβO and the resultant neuronal cell death. These cAMP protective effects are dependent on protein kinase A, but not dependent on the exchange protein directly activated by cAMP. Conceivably, cAMP protects neuronal cells from AβO-induced cytotoxicity by decreasing cell-surface-associated PrPC and 67LR.
Collapse
Affiliation(s)
- Rayudu Gopalakrishna
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA,Corresponding author: Department of Integrative Anatomical Sciences, 1333 San Pablo Street, Keck School of Medicine, Los Angeles, CA 90089, USA, Phone: 1 + 323-442-1770; Fax: 1 + 323-442-1771:
| | - Charlotte Y. Lin
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrew Oh
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Calvin Le
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Seolyn Yang
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Alexandra Hicks
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Mark S. Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA; James A. Haley VA Medical Center, Tampa, FL 33612, USA
| | - William J. Mack
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Narayan R. Bhat
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
29
|
Marmolejo-Garza A, Medeiros-Furquim T, Rao R, Eggen BJL, Boddeke E, Dolga AM. Transcriptomic and epigenomic landscapes of Alzheimer's disease evidence mitochondrial-related pathways. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119326. [PMID: 35839870 DOI: 10.1016/j.bbamcr.2022.119326] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 02/06/2023]
Abstract
Alzheimers disease (AD) is the main cause of dementia and it is defined by cognitive decline coupled to extracellular deposit of amyloid-beta protein and intracellular hyperphosphorylation of tau protein. Historically, efforts to target such hallmarks have failed in numerous clinical trials. In addition to these hallmark-targeted approaches, several clinical trials focus on other AD pathological processes, such as inflammation, mitochondrial dysfunction, and oxidative stress. Mitochondria and mitochondrial-related mechanisms have become an attractive target for disease-modifying strategies, as mitochondrial dysfunction prior to clinical onset has been widely described in AD patients and AD animal models. Mitochondrial function relies on both the nuclear and mitochondrial genome. Findings from omics technologies have shed light on AD pathophysiology at different levels (e.g., epigenome, transcriptome and proteome). Most of these studies have focused on the nuclear-encoded components. The first part of this review provides an updated overview of the mechanisms that regulate mitochondrial gene expression and function. The second part of this review focuses on evidence of mitochondrial dysfunction in AD. We have focused on published findings and datasets that study AD. We analyzed published data and provide examples for mitochondrial-related pathways. These pathways are strikingly dysregulated in AD neurons and glia in sex-, cell- and disease stage-specific manners. Analysis of mitochondrial omics data highlights the involvement of mitochondria in AD, providing a rationale for further disease modeling and drug targeting.
Collapse
Affiliation(s)
- Alejandro Marmolejo-Garza
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, the Netherlands; Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tiago Medeiros-Furquim
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, the Netherlands; Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ramya Rao
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, the Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Erik Boddeke
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen N, Denmark.
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, the Netherlands.
| |
Collapse
|
30
|
Ryan KC, Laboy JT, Norman KR. Deregulation of Mitochondrial Calcium Handling Due to Presenilin Loss Disrupts Redox Homeostasis and Promotes Neuronal Dysfunction. Antioxidants (Basel) 2022; 11:antiox11091642. [PMID: 36139715 PMCID: PMC9495597 DOI: 10.3390/antiox11091642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dysfunction and oxidative stress are major contributors to the pathophysiology of neurodegenerative diseases, including Alzheimer’s disease (AD). However, the mechanisms driving mitochondrial dysfunction and oxidative stress are unclear. Familial AD (fAD) is an early onset form of AD caused primarily by mutations in the presenilin-encoding genes. Previously, using Caenorhabditis elegans as a model system to study presenilin function, we found that loss of C. elegans presenilin orthologue SEL-12 results in elevated mitochondrial and cytosolic calcium levels. Here, we provide evidence that elevated neuronal mitochondrial generated reactive oxygen species (ROS) and subsequent neurodegeneration in sel-12 mutants are a consequence of the increase of mitochondrial calcium levels and not cytosolic calcium levels. We also identify mTORC1 signaling as a critical factor in sustaining high ROS in sel-12 mutants in part through its repression of the ROS scavenging system SKN-1/Nrf. Our study reveals that SEL-12/presenilin loss disrupts neuronal ROS homeostasis by increasing mitochondrial ROS generation and elevating mTORC1 signaling, which exacerbates this imbalance by suppressing SKN-1/Nrf antioxidant activity.
Collapse
|
31
|
Adeoye T, Shah SI, Demuro A, Rabson DA, Ullah G. Upregulated Ca 2+ Release from the Endoplasmic Reticulum Leads to Impaired Presynaptic Function in Familial Alzheimer's Disease. Cells 2022; 11:2167. [PMID: 35883609 PMCID: PMC9315668 DOI: 10.3390/cells11142167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 12/10/2022] Open
Abstract
Neurotransmitter release from presynaptic terminals is primarily regulated by rapid Ca2+ influx through membrane-resident voltage-gated Ca2+ channels (VGCCs). Moreover, accumulating evidence indicates that the endoplasmic reticulum (ER) is extensively present in axonal terminals of neurons and plays a modulatory role in synaptic transmission by regulating Ca2+ levels. Familial Alzheimer's disease (FAD) is marked by enhanced Ca2+ release from the ER and downregulation of Ca2+ buffering proteins. However, the precise consequence of impaired Ca2+ signaling within the vicinity of VGCCs (active zone (AZ)) on exocytosis is poorly understood. Here, we perform in silico experiments of intracellular Ca2+ signaling and exocytosis in a detailed biophysical model of hippocampal synapses to investigate the effect of aberrant Ca2+ signaling on neurotransmitter release in FAD. Our model predicts that enhanced Ca2+ release from the ER increases the probability of neurotransmitter release in FAD. Moreover, over very short timescales (30-60 ms), the model exhibits activity-dependent and enhanced short-term plasticity in FAD, indicating neuronal hyperactivity-a hallmark of the disease. Similar to previous observations in AD animal models, our model reveals that during prolonged stimulation (~450 ms), pathological Ca2+ signaling increases depression and desynchronization with stimulus, causing affected synapses to operate unreliably. Overall, our work provides direct evidence in support of a crucial role played by altered Ca2+ homeostasis mediated by intracellular stores in FAD.
Collapse
Affiliation(s)
- Temitope Adeoye
- Department of Physics, University of South Florida, Tampa, FL 33620, USA; (T.A.); (S.I.S.); (D.A.R.)
| | - Syed I. Shah
- Department of Physics, University of South Florida, Tampa, FL 33620, USA; (T.A.); (S.I.S.); (D.A.R.)
| | - Angelo Demuro
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA;
| | - David A. Rabson
- Department of Physics, University of South Florida, Tampa, FL 33620, USA; (T.A.); (S.I.S.); (D.A.R.)
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620, USA; (T.A.); (S.I.S.); (D.A.R.)
| |
Collapse
|
32
|
Zernov N, Bezprozvanny I, Popugaeva E. CaMKIIβ knockdown decreases store-operated calcium entry in hippocampal dendritic spines. IBRO Neurosci Rep 2022; 12:90-97. [PMID: 35079728 PMCID: PMC8777283 DOI: 10.1016/j.ibneur.2022.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/06/2022] [Accepted: 01/08/2022] [Indexed: 01/13/2023] Open
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) and neuronal store-operated calcium entry (nSOCE) have been implicated in the development of Alzheimer's disease (AD). nSOCE is involved in regulation of dendritic spine shape, particularly in stability of mushroom spines that play role in formation of strong synapses. CaMKII is involved in regulation of induction of long-term potentiation, that is needed for shaping of memory. In the present study, we demonstrated that inhibition of kinase activity of CaMKII by KN-62 decreases nSOCE amplitude in soma of primary hippocampal neurons. We have shown that knockdown of CaMKIIβ leads to the downregulation of nSOCE in dendritic spines. In agreement with previously published data, we have also observed that CaMKIIβ knockdown causes mushroom spine loss in primary hippocampal culture. The effect of CaMKIIβ knockdown on the nSOCE may be associated with a decrease of dendritic spine head size.
Collapse
Affiliation(s)
- Nikita Zernov
- Peter the Great St.Petersburg Polytechnic University, Laboratory of Molecular Neurodegeneration, St.Petersburg, Russia
| | - Ilya Bezprozvanny
- Peter the Great St.Petersburg Polytechnic University, Laboratory of Molecular Neurodegeneration, St.Petersburg, Russia
- UT Southwestern Medical Center, Department of Physiology, Dallas, USA
| | - Elena Popugaeva
- Peter the Great St.Petersburg Polytechnic University, Laboratory of Molecular Neurodegeneration, St.Petersburg, Russia
- Corresponding author.
| |
Collapse
|
33
|
Zhang B, Chen T, Cao M, Yuan C, Reiter RJ, Zhao Z, Zhao Y, Chen L, Fan W, Wang X, Zhou X, Li C. Gut Microbiota Dysbiosis Induced by Decreasing Endogenous Melatonin Mediates the Pathogenesis of Alzheimer's Disease and Obesity. Front Immunol 2022; 13:900132. [PMID: 35619714 PMCID: PMC9127079 DOI: 10.3389/fimmu.2022.900132] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/13/2022] [Indexed: 01/15/2023] Open
Abstract
Lifestyle choices, external environment, aging, and other factors influence the synthesis of melatonin. Although the physiological functions of melatonin have been widely studied in relation to specific organs, the systemic effects of endogenous melatonin reduction has not been reported. This study evaluates the systemic changes and possible pathogenic risks in an endogenous melatonin reduction (EMR) mouse model deficient in the rate limiting enzyme in melatonin production, arylalkylamine N-acetyltransferase (Aanat) gene. Using this model, we identified a new relationship between melatonin, Alzheimer’s disease (AD), and gut microbiota. Systematic changes were evaluated using multi-omics analysis. Fecal microbiota transplantation (FMT) was performed to examine the role of gut microbiota in the pathogenic risks of EMR. EMR mice exhibited a pan-metabolic disorder, with significant transcriptome changes in 11 organs, serum metabolome alterations as well as microbiota dysbiosis. Microbiota dysbiosis was accompanied by increased gut permeability along with gut and systemic inflammation. Correlation analysis revealed that systemic inflammation may be related to the increase of Ruminiclostridium_5 relative abundance. 8-month-old EMR mice had AD-like phenotypes, including Iba-1 activation, A β protein deposition and decreased spatial memory ability. Moreover, EMR mice showed decreased anti stress ability, under high-fat diet, EMR mice had greater body weight and more obvious hepatic steatosis compared with WT group. FMT improved gut permeability, systemic inflammation, and AD-related phenotypes, while reducing obesity in EMR mice. Our findings suggest EMR causes systemic changes mediated by gut microbiota dysbiosis, which may be a pathogenic factor for AD and obesity, we further proved the gut microbiota is a potential target for the prevention and treatment of AD and obesity.
Collapse
Affiliation(s)
- Boqi Zhang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Tong Chen
- College of Animal Sciences, Jilin University, Changchun, China
| | - Maosheng Cao
- College of Animal Sciences, Jilin University, Changchun, China
| | - Chenfeng Yuan
- College of Animal Sciences, Jilin University, Changchun, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Zijiao Zhao
- College of Animal Sciences, Jilin University, Changchun, China
| | - Yun Zhao
- College of Animal Sciences, Jilin University, Changchun, China
| | - Lu Chen
- College of Animal Sciences, Jilin University, Changchun, China
| | - Wenjing Fan
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xin Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xu Zhou
- College of Animal Sciences, Jilin University, Changchun, China
| | - Chunjin Li
- College of Animal Sciences, Jilin University, Changchun, China
| |
Collapse
|
34
|
Kim S, Kim DK, Jeong S, Lee J. The Common Cellular Events in the Neurodegenerative Diseases and the Associated Role of Endoplasmic Reticulum Stress. Int J Mol Sci 2022; 23:5894. [PMID: 35682574 PMCID: PMC9180188 DOI: 10.3390/ijms23115894] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022] Open
Abstract
Neurodegenerative diseases are inseparably linked with aging and increase as life expectancy extends. There are common dysfunctions in various cellular events shared among neurogenerative diseases, such as calcium dyshomeostasis, neuroinflammation, and age-associated decline in the autophagy-lysosome system. However, most of all, the prominent pathological feature of neurodegenerative diseases is the toxic buildup of misfolded protein aggregates and inclusion bodies accompanied by an impairment in proteostasis. Recent studies have suggested a close association between endoplasmic reticulum (ER) stress and neurodegenerative pathology in cellular and animal models as well as in human patients. The contribution of mutant or misfolded protein-triggered ER stress and its associated signaling events, such as unfolded protein response (UPR), to the pathophysiology of various neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's disease, amyotrophic lateral sclerosis, and prion disease, is described here. Impaired UPR action is commonly attributed to exacerbated ER stress, pathogenic protein aggregate accumulation, and deteriorating neurodegenerative pathologies. Thus, activating certain UPR components has been shown to alleviate ER stress and its associated neurodegeneration. However, uncontrolled activation of some UPR factors has also been demonstrated to worsen neurodegenerative phenotypes, suggesting that detailed molecular mechanisms around ER stress and its related neurodegenerations should be understood to develop effective therapeutics against aging-associated neurological syndromes. We also discuss current therapeutic endeavors, such as the development of small molecules that selectively target individual UPR components and address ER stress in general.
Collapse
Affiliation(s)
- Soojeong Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
| | - Doo Kyung Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
| | - Seho Jeong
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| |
Collapse
|
35
|
Sugiura A, Kitamura M, Hasegawa Y. Calcium carbonate supplementation causes motor dysfunction. Exp Anim 2022; 71:399-410. [PMID: 35584940 PMCID: PMC9388334 DOI: 10.1538/expanim.22-0011] [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] [Indexed: 11/27/2022] Open
Abstract
We previously showed that a diet containing calcium carbonate causes impairments in spatial and recognition memory in mice. In this study, we investigated the effects of calcium carbonate
supplementation on motor function. Motor function was determined using different tests that have been used to analyze different aspects of Parkinsonism. A catalepsy test for akinesia; a
muscular strength assessment, pole test, beam-walking test, and gait analysis for motor coordination and balance assessment; and an open-field test for locomotor activity assessment were
performed. The mice were fed diets containing 0.6% or 1.0% calcium carbonate for eight weeks, after which they were evaluated for motor functions. The diets containing calcium carbonate
caused significant motor dysfunction, as revealed by the different tests, although the spontaneous locomotor activity did not change. Calcium carbonate supplementation decreased the dopamine
content in the basal ganglia, including the striatum and substantia nigra, and the number of tyrosine hydroxylase-positive neurons in the substantia nigra. In addition, administration of
L-dopa led to at least a partial recovery of motor dysfunction, suggesting that calcium carbonate supplementation causes motor dysfunction by decreasing the dopamine content in the basal
ganglia. These results suggest that mice with calcium carbonate-induced motor dysfunction may be useful as a new animal model for Parkinson’s disease and Huntington’s disease.
Collapse
Affiliation(s)
- Ami Sugiura
- College of Environmental Technology, Muroran Institute of Technology
| | - Misa Kitamura
- College of Environmental Technology, Muroran Institute of Technology
| | - Yasushi Hasegawa
- College of Environmental Technology, Muroran Institute of Technology
| |
Collapse
|
36
|
Hnath B, Dokholyan NV. Toxic SOD1 trimers are off-pathway in the formation of amyloid-like fibrils in ALS. Biophys J 2022; 121:2084-2095. [PMID: 35505609 DOI: 10.1016/j.bpj.2022.04.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 04/01/2022] [Accepted: 04/28/2022] [Indexed: 11/26/2022] Open
Abstract
Accumulation of insoluble amyloid fibrils is widely studied as a critical factor in the pathology of multiple neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease. Misfolded Cu, Zn superoxide dismutase (SOD1) was the first protein linked to ALS, and non-native SOD1 trimeric oligomers were recently linked to cytotoxicity, while larger oligomers were protective to cells. The balance between trimers and larger aggregates in the process of SOD1 aggregation is, thus, a critical determinant of potential therapeutic approaches to treat ALS. Yet, it is unknown whether these trimeric oligomers are a necessary intermediate for larger aggregate formation or a distinct off-pathway species competing with fibril formation. Depending on the on- or off-pathway scenario of trimer formation, we expect drastically different therapeutic approaches. Here, we show that the toxic SOD1 trimer is an off-pathway intermediate competing with protective fibril formation. We design mutant SOD1 constructs that remain in a trimeric state (super stable trimers) and show that stabilizing the trimeric SOD1 prevents formation of fibrils in vitro and in a motor neuron like cell model (NSC-34). Using size exclusion chromatography, we track the aggregation kinetics of purified SOD1 and show direct competition of trimeric SOD1 with larger oligomer and fibril formation. Finally, we show the trimer is structurally independent of both larger soluble oligomers and insoluble fibrils using circular dichroism spectroscopy and limited proteolysis.
Collapse
Affiliation(s)
- Brianna Hnath
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
37
|
Park J, Hee Kim S, Kim YJ, Kim H, Oh Y, Yeong Choi K, Kim BC, Ho Lee K, Keun Song W. Elevation of phospholipase C-β1 expression by amyloid-β facilitates calcium overload in neuronal cells. Brain Res 2022; 1788:147924. [DOI: 10.1016/j.brainres.2022.147924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/26/2022] [Accepted: 04/19/2022] [Indexed: 11/02/2022]
|
38
|
Garbincius JF, Elrod JW. Mitochondrial calcium exchange in physiology and disease. Physiol Rev 2022; 102:893-992. [PMID: 34698550 PMCID: PMC8816638 DOI: 10.1152/physrev.00041.2020] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 08/16/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
The uptake of calcium into and extrusion of calcium from the mitochondrial matrix is a fundamental biological process that has critical effects on cellular metabolism, signaling, and survival. Disruption of mitochondrial calcium (mCa2+) cycling is implicated in numerous acquired diseases such as heart failure, stroke, neurodegeneration, diabetes, and cancer and is genetically linked to several inherited neuromuscular disorders. Understanding the mechanisms responsible for mCa2+ exchange therefore holds great promise for the treatment of these diseases. The past decade has seen the genetic identification of many of the key proteins that mediate mitochondrial calcium uptake and efflux. Here, we present an overview of the phenomenon of mCa2+ transport and a comprehensive examination of the molecular machinery that mediates calcium flux across the inner mitochondrial membrane: the mitochondrial uniporter complex (consisting of MCU, EMRE, MICU1, MICU2, MICU3, MCUB, and MCUR1), NCLX, LETM1, the mitochondrial ryanodine receptor, and the mitochondrial permeability transition pore. We then consider the physiological implications of mCa2+ flux and evaluate how alterations in mCa2+ homeostasis contribute to human disease. This review concludes by highlighting opportunities and challenges for therapeutic intervention in pathologies characterized by aberrant mCa2+ handling and by summarizing critical unanswered questions regarding the biology of mCa2+ flux.
Collapse
Affiliation(s)
- Joanne F Garbincius
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
39
|
Zhang R, Zhang Y, Guo F, Li S, Cui H. RNA N6-Methyladenosine Modifications and Its Roles in Alzheimer's Disease. Front Cell Neurosci 2022; 16:820378. [PMID: 35401117 PMCID: PMC8989074 DOI: 10.3389/fncel.2022.820378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
The importance of epitranscriptomics in regulating gene expression has received widespread attention. Recently, RNA methylation modifications, particularly N6-methyladenosine (m6A), have received marked attention. m6A, the most common and abundant type of eukaryotic methylation modification in RNAs, is a dynamic reversible modification that regulates nuclear splicing, stability, translation, and subcellular localization of RNAs. These processes are involved in the occurrence and development of many diseases. An increasing number of studies have focused on the role of m6A modification in Alzheimer's disease, which is the most common neurodegenerative disease. This review focuses on the general features, mechanisms, and functions of m6A methylation modification and its role in Alzheimer's disease.
Collapse
Affiliation(s)
- Runjiao Zhang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Yizhou Zhang
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Fangzhen Guo
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Sha Li
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Huixian Cui
- Department of Anatomy, Hebei Medical University, Shijiazhuang, China
- Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| |
Collapse
|
40
|
CaMKIIα Signaling Is Required for the Neuroprotective Effects of Dl-3-n-Butylphthalide in Alzheimer's Disease. Mol Neurobiol 2022; 59:3370-3381. [PMID: 35305243 DOI: 10.1007/s12035-022-02777-8] [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: 10/21/2021] [Accepted: 02/15/2022] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative disease and most anti-AD drugs have failed in clinical trials; hence, it is urgent to find potentially effective drugs against AD. DL-3-n-butylphthalide (NBP) is a compound extracted from celery seed and is a multiple-target drug. Several studies have demonstrated the neuroprotective effects of NBP on cognitive impairment, but the mechanisms of NBP remains relatively unexplored. In this study, we found that NBP could alleviated the increase of intracellular Ca2+ and reversed down-regulation of Ca2+/calmodulin-dependent protein kinase alpha (CaMKIIα) signaling and rescued neuronal apoptosis in SH-SY5Y cells treated by Aβ oligomers. However, these neuroprotective effects of NBP on neuronal damage and CaMKIIα signaling were abolished when CaMKIIα expression was knocked down or its activity was inhibited. Thus, our findings suggested that CaMKIIα signaling was required for the neuroprotective effects of NBP in AD and provided an improved basis for elucidating the mechanism and treatment of NBP in AD.
Collapse
|
41
|
Parys JB, Bultynck G. Preface to the Special Issue of the European Calcium Society in honor of Professor Sir Michael J. Berridge. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119172. [PMID: 34774604 DOI: 10.1016/j.bbamcr.2021.119172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Jan B Parys
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, BE-3000 Leuven, Belgium.
| | - Geert Bultynck
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, BE-3000 Leuven, Belgium.
| |
Collapse
|
42
|
Hu F, Song X, Long D. Transient receptor potential ankyrin 1 and calcium: Interactions and association with disease (Review). Exp Ther Med 2021; 22:1462. [PMID: 34737802 PMCID: PMC8561754 DOI: 10.3892/etm.2021.10897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
Calcium (Ca2+) is an essential signaling molecule in all cells. It is involved in numerous fundamental functions, including cell life and death. Abnormal regulation of Ca2+ homeostasis may cause human diseases. Usually known as a member of the transient receptor potential (TRP) family, TRP ankyrin 1 (TRPA1) is the only member of the ankyrin subfamily identified in mammals so far and widely expressed in cells and tissues. As it is involved in numerous sensory disorders such as pain and pruritus, TRPA1 is a potential target for the treatment of neuropathy. The functions of TRP family members are closely related to Ca2+. TRPA1 has a high permeability to Ca2+, sodium and potassium ions as a non-selective cation channel and the Ca2+ influx mediated by TRPA1 is involved in a variety of biological processes. In the present review, research on the relationship between the TRPA1 channel and Ca2+ ions and their interaction in disease-associated processes was summarised. The therapeutic potential of the TRPA1 channel is highlighted, which is expected to become a novel direction for the prevention and treatment of health conditions such as cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Fangyan Hu
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiaohua Song
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Dingxin Long
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
43
|
Gerasimov E, Erofeev A, Borodinova A, Bolshakova A, Balaban P, Bezprozvanny I, Vlasova OL. Optogenetic Activation of Astrocytes-Effects on Neuronal Network Function. Int J Mol Sci 2021; 22:9613. [PMID: 34502519 PMCID: PMC8431749 DOI: 10.3390/ijms22179613] [Citation(s) in RCA: 13] [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: 07/28/2021] [Revised: 08/24/2021] [Accepted: 09/01/2021] [Indexed: 01/04/2023] Open
Abstract
Optogenetics approach is used widely in neurobiology as it allows control of cellular activity with high spatial and temporal resolution. In most studies, optogenetics is used to control neuronal activity. In the present study optogenetics was used to stimulate astrocytes with the aim to modulate neuronal activity. To achieve this goal, light stimulation was applied to astrocytes expressing a version of ChR2 (ionotropic opsin) or Opto-α1AR (metabotropic opsin). Optimal optogenetic stimulation parameters were determined using patch-clamp recordings of hippocampal pyramidal neurons' spontaneous activity in brain slices as a readout. It was determined that the greatest increase in the number of spontaneous synaptic currents was observed when astrocytes expressing ChR2(H134R) were activated by 5 s of continuous light. For the astrocytes expressing Opto-α1AR, the greatest response was observed in the pulse stimulation mode (T = 1 s, t = 100 ms). It was also observed that activation of the astrocytic Opto-a1AR but not ChR2 results in an increase of the fEPSP slope in hippocampal neurons. Based on these results, we concluded that Opto-a1AR expressed in hippocampal astrocytes provides an opportunity to modulate the long-term synaptic plasticity optogenetically, and may potentially be used to normalize the synaptic transmission and plasticity defects in a variety of neuropathological conditions, including models of Alzheimer's disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Evgenii Gerasimov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (A.E.); (A.B.); (I.B.)
| | - Alexander Erofeev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (A.E.); (A.B.); (I.B.)
| | - Anastasia Borodinova
- Cellular Neurobiology of Learning Lab, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Science, Butlerova St. 5A, 117485 Moscow, Russia; (A.B.); (P.B.)
| | - Anastasia Bolshakova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (A.E.); (A.B.); (I.B.)
| | - Pavel Balaban
- Cellular Neurobiology of Learning Lab, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Science, Butlerova St. 5A, 117485 Moscow, Russia; (A.B.); (P.B.)
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (A.E.); (A.B.); (I.B.)
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Olga L. Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Khlopina St. 11, 194021 St. Petersburg, Russia; (A.E.); (A.B.); (I.B.)
| |
Collapse
|
44
|
Wu S, Yang M, Kim P, Zhou X. ADeditome provides the genomic landscape of A-to-I RNA editing in Alzheimer's disease. Brief Bioinform 2021; 22:bbaa384. [PMID: 33401309 PMCID: PMC8424397 DOI: 10.1093/bib/bbaa384] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/08/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
A-to-I RNA editing, contributing to nearly 90% of all editing events in human, has been reported to involve in the pathogenesis of Alzheimer's disease (AD) due to its roles in brain development and immune regulation, such as the deficient editing of GluA2 Q/R related to cell death and memory loss. Currently, there are urgent needs for the systematic annotations of A-to-I RNA editing events in AD. Here, we built ADeditome, the annotation database of A-to-I RNA editing in AD available at https://ccsm.uth.edu/ADeditome, aiming to provide a resource and reference for functional annotation of A-to-I RNA editing in AD to identify therapeutically targetable genes in an individual. We detected 1676 363 editing sites in 1524 samples across nine brain regions from ROSMAP, MayoRNAseq and MSBB. For these editing events, we performed multiple functional annotations including identification of specific and disease stage associated editing events and the influence of editing events on gene expression, protein recoding, alternative splicing and miRNA regulation for all the genes, especially for AD-related genes in order to explore the pathology of AD. Combing all the analysis results, we found 108 010 and 26 168 editing events which may promote or inhibit AD progression, respectively. We also found 5582 brain region-specific editing events with potentially dual roles in AD across different brain regions. ADeditome will be a unique resource for AD and drug research communities to identify therapeutically targetable editing events. Significance: ADeditome is the first comprehensive resource of the functional genomics of individual A-to-I RNA editing events in AD, which will be useful for many researchers in the fields of AD pathology, precision medicine, and therapeutic researches.
Collapse
Affiliation(s)
- Sijia Wu
- School of Life Science and Technology, Xidian University, Xi'an, China
| | | | | | | |
Collapse
|
45
|
Gadhave K, Kumar D, Uversky VN, Giri R. A multitude of signaling pathways associated with Alzheimer's disease and their roles in AD pathogenesis and therapy. Med Res Rev 2021; 41:2689-2745. [PMID: 32783388 PMCID: PMC7876169 DOI: 10.1002/med.21719] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/13/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
The exact molecular mechanisms associated with Alzheimer's disease (AD) pathology continue to represent a mystery. In the past decades, comprehensive data were generated on the involvement of different signaling pathways in the AD pathogenesis. However, the utilization of signaling pathways as potential targets for the development of drugs against AD is rather limited due to the immense complexity of the brain and intricate molecular links between these pathways. Therefore, finding a correlation and cross-talk between these signaling pathways and establishing different therapeutic targets within and between those pathways are needed for better understanding of the biological events responsible for the AD-related neurodegeneration. For example, autophagy is a conservative cellular process that shows link with many other AD-related pathways and is crucial for maintenance of the correct cellular balance by degrading AD-associated pathogenic proteins. Considering the central role of autophagy in AD and its interplay with many other pathways, the finest therapeutic strategy to fight against AD is the use of autophagy as a target. As an essential step in this direction, this comprehensive review represents recent findings on the individual AD-related signaling pathways, describes key features of these pathways and their cross-talk with autophagy, represents current drug development, and introduces some of the multitarget beneficial approaches and strategies for the therapeutic intervention of AD.
Collapse
Affiliation(s)
- Kundlik Gadhave
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Deepak Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Vladimir N. Uversky
- Department of Molecular Medicine and Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| |
Collapse
|
46
|
Integrated Bioinformatics Analysis to Identify Alternative Therapeutic Targets for Alzheimer's Disease: Insights from a Synaptic Machinery Perspective. J Mol Neurosci 2021; 72:273-286. [PMID: 34414562 DOI: 10.1007/s12031-021-01893-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD), the most common type of dementia, is a serious neurodegenerative disease that has no cure yet, but whose symptoms can be alleviated with available medications. Therefore, early and accurate diagnosis of the disease and elucidation of the molecular mechanisms involved in the progression of pathogenesis are critically important. This study aimed to identify dysregulated miRNAs and their target mRNAs through the integrated analysis of miRNA and mRNA expression profiling in AD patients versus unaffected controls. Expression profiles in postmortem brain samples from AD patients and healthy individuals were extracted from the Gene Expression Omnibus database and were analyzed using bioinformatics approaches to identify gene ontologies, pathways, and networks. Finally, the module analysis of the PPI network and hub gene selection was carried out. A total of five differentially expressed miRNAs were extracted from the miRNA dataset, and 4312 differentially expressed mRNAs were obtained from the mRNA dataset. By comparing the DEGs and the putative targets of the altered miRNAs, 116 (3 upregulated and 113 downregulated) coordinated genes were determined. Also, six hub genes (SNAP25, GRIN2A, GRIN2B, DLG2, ATP2B2, and SCN2A) were identified by constructing a PPI network. The results of the present study provide insight into mechanisms such as synaptic machinery and neuronal communication underlying AD pathogenesis, specifically concerning miRNAs.
Collapse
|
47
|
Wang F, Xu CS, Chen WH, Duan SW, Xu SJ, Dai JJ, Wang QW. Identification of Blood-Based Glycolysis Gene Associated with Alzheimer's Disease by Integrated Bioinformatics Analysis. J Alzheimers Dis 2021; 83:163-178. [PMID: 34308907 DOI: 10.3233/jad-210540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is one of many common neurodegenerative diseases without ideal treatment, but early detection and intervention can prevent the disease progression. OBJECTIVE This study aimed to identify AD-related glycolysis gene for AD diagnosis and further investigation by integrated bioinformatics analysis. METHODS 122 subjects were recruited from the affiliated hospitals of Ningbo University between 1 October 2015 and 31 December 2016. Their clinical information and methylation levels of 8 glycolysis genes were assessed. Machine learning algorithms were used to establish an AD prediction model. Receiver operating characteristic curve (AUC) and decision curve analysis (DCA) were used to assess the model. An AD risk factor model was developed by SHapley Additive exPlanations (SHAP) to extract features that had important impacts on AD. Finally, gene expression of AD-related glycolysis genes were validated by AlzData. RESULTS An AD prediction model was developed using random forest algorithm with the best average ROC_AUC (0.969544). The threshold probability of the model was positive in the range of 0∼0.9875 by DCA. Eight glycolysis genes (GAPDHS, PKLR, PFKFB3, LDHC, DLD, ALDOC, LDHB, HK3) were identified by SHAP. Five of these genes (PFKFB3, DLD, ALDOC, LDHB, LDHC) have significant differences in gene expression between AD and control groups by Alzdata, while three of the genes (HK3, ALDOC, PKLR) are related to the pathogenesis of AD. GAPDHS is involved in the regulatory network of AD risk genes. CONCLUSION We identified 8 AD-related glycolysis genes (GAPDHS, PFKFB3, LDHC, HK3, ALDOC, LDHB, PKLR, DLD) as promising candidate biomarkers for early diagnosis of AD by integrated bioinformatics analysis. Machine learning has the advantage in identifying genes.
Collapse
Affiliation(s)
- Fng Wang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China.,Zhejiang Pharmaceutical College, Ningbo, China
| | - Chun-Shuang Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Wei-Hua Chen
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Shi-Wei Duan
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Shu-Jun Xu
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Jun-Jie Dai
- Affiliated Lihuili Hospital of Ningbo University, Ningbo, China
| | - Qin-Wen Wang
- Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
48
|
Jadiya P, Garbincius JF, Elrod JW. Reappraisal of metabolic dysfunction in neurodegeneration: Focus on mitochondrial function and calcium signaling. Acta Neuropathol Commun 2021; 9:124. [PMID: 34233766 PMCID: PMC8262011 DOI: 10.1186/s40478-021-01224-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
The cellular and molecular mechanisms that drive neurodegeneration remain poorly defined. Recent clinical trial failures, difficult diagnosis, uncertain etiology, and lack of curative therapies prompted us to re-examine other hypotheses of neurodegenerative pathogenesis. Recent reports establish that mitochondrial and calcium dysregulation occur early in many neurodegenerative diseases (NDDs), including Alzheimer's disease, Parkinson's disease, Huntington's disease, and others. However, causal molecular evidence of mitochondrial and metabolic contributions to pathogenesis remains insufficient. Here we summarize the data supporting the hypothesis that mitochondrial and metabolic dysfunction result from diverse etiologies of neuropathology. We provide a current and comprehensive review of the literature and interpret that defective mitochondrial metabolism is upstream and primary to protein aggregation and other dogmatic hypotheses of NDDs. Finally, we identify gaps in knowledge and propose therapeutic modulation of mCa2+ exchange and mitochondrial function to alleviate metabolic impairments and treat NDDs.
Collapse
Affiliation(s)
- Pooja Jadiya
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad St, MERB 949, Philadelphia, PA, 19140, USA
| | - Joanne F Garbincius
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad St, MERB 949, Philadelphia, PA, 19140, USA
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad St, MERB 949, Philadelphia, PA, 19140, USA.
| |
Collapse
|
49
|
Alves SS, Silva-Junior RMPD, Servilha-Menezes G, Homolak J, Šalković-Petrišić M, Garcia-Cairasco N. Insulin Resistance as a Common Link Between Current Alzheimer's Disease Hypotheses. J Alzheimers Dis 2021; 82:71-105. [PMID: 34024838 DOI: 10.3233/jad-210234] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Almost 115 years ago, Alois Alzheimer described Alzheimer's disease (AD) for the first time. Since then, many hypotheses have been proposed. However, AD remains a severe health public problem. The current medical approaches for AD are limited to symptomatic interventions and the complexity of this disease has led to a failure rate of approximately 99.6%in AD clinical trials. In fact, no new drug has been approved for AD treatment since 2003. These failures indicate that we are failing in mimicking this disease in experimental models. Although most studies have focused on the amyloid cascade hypothesis of AD, the literature has made clear that AD is rather a multifactorial disorder. Therefore, the persistence in a single theory has resulted in lost opportunities. In this review, we aim to present the striking points of the long scientific path followed since the description of the first AD case and the main AD hypotheses discussed over the last decades. We also propose insulin resistance as a common link between many other hypotheses.
Collapse
Affiliation(s)
- Suélen Santos Alves
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Rui Milton Patrício da Silva-Junior
- Department of Internal Medicine, Ribeirão Preto Medical School -University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Gabriel Servilha-Menezes
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Melita Šalković-Petrišić
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
50
|
Chanaday NL, Nosyreva E, Shin OH, Zhang H, Aklan I, Atasoy D, Bezprozvanny I, Kavalali ET. Presynaptic store-operated Ca 2+ entry drives excitatory spontaneous neurotransmission and augments endoplasmic reticulum stress. Neuron 2021; 109:1314-1332.e5. [PMID: 33711258 PMCID: PMC8068669 DOI: 10.1016/j.neuron.2021.02.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 01/18/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022]
Abstract
Store-operated calcium entry (SOCE) is activated by depletion of Ca2+ from the endoplasmic reticulum (ER) and mediated by stromal interaction molecule (STIM) proteins. Here, we show that in rat and mouse hippocampal neurons, acute ER Ca2+ depletion increases presynaptic Ca2+ levels and glutamate release through a pathway dependent on STIM2 and the synaptic Ca2+ sensor synaptotagmin-7 (syt7). In contrast, synaptotagmin-1 (syt1) can suppress SOCE-mediated spontaneous release, and STIM2 is required for the increase in spontaneous release seen during syt1 loss of function. We also demonstrate that chronic ER stress activates the same pathway leading to syt7-dependent potentiation of spontaneous glutamate release. During ER stress, inhibition of SOCE or syt7-driven fusion partially restored basal neurotransmission and decreased expression of pro-apoptotic markers, indicating that these processes participate in the amplification of ER-stress-related damage. Taken together, we propose that presynaptic SOCE links ER stress and augmented spontaneous neurotransmission, which may, in turn, facilitate neurodegeneration.
Collapse
Affiliation(s)
- Natali L. Chanaday
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240-7933, USA
| | - Elena Nosyreva
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Ok-Ho Shin
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240-7933, USA
| | - Hua Zhang
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA
| | - Iltan Aklan
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA,FOE Diabetes Research Center, University of Iowa, Iowa City, IA, 52242, USA
| | - Ilya Bezprozvanny
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA.,Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg State Polytechnic University, St. Petersburg, Russia
| | - Ege T. Kavalali
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, 37240-7933, USA.,Vanderbilt Brain Institute.,Corresponding author: Ege T. Kavalali, Ph.D., Department of Pharmacology, Vanderbilt University, 465 21st Avenue South, 7130A MRBIII, PMB407933 Nashville, TN 37240-7933, phone: 615-343-5480,
| |
Collapse
|