1
|
Woo MS, Engler JB, Friese MA. The neuropathobiology of multiple sclerosis. Nat Rev Neurosci 2024; 25:493-513. [PMID: 38789516 DOI: 10.1038/s41583-024-00823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
Chronic low-grade inflammation and neuronal deregulation are two components of a smoldering disease activity that drives the progression of disability in people with multiple sclerosis (MS). Although several therapies exist to dampen the acute inflammation that drives MS relapses, therapeutic options to halt chronic disability progression are a major unmet clinical need. The development of such therapies is hindered by our limited understanding of the neuron-intrinsic determinants of resilience or vulnerability to inflammation. In this Review, we provide a neuron-centric overview of recent advances in deciphering neuronal response patterns that drive the pathology of MS. We describe the inflammatory CNS environment that initiates neurotoxicity by imposing ion imbalance, excitotoxicity and oxidative stress, and by direct neuro-immune interactions, which collectively lead to mitochondrial dysfunction and epigenetic dysregulation. The neuronal demise is further amplified by breakdown of neuronal transport, accumulation of cytosolic proteins and activation of cell death pathways. Continuous neuronal damage perpetuates CNS inflammation by activating surrounding glia cells and by directly exerting toxicity on neighbouring neurons. Further, we explore strategies to overcome neuronal deregulation in MS and compile a selection of neuronal actuators shown to impact neurodegeneration in preclinical studies. We conclude by discussing the therapeutic potential of targeting such neuronal actuators in MS, including some that have already been tested in interventional clinical trials.
Collapse
Affiliation(s)
- Marcel S Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
2
|
Jos S, Poulose R, Kambaru A, Gogoi H, Dalavaikodihalli Nanjaiah N, Padmanabhan B, Mehta B, Padavattan S. Tau-S214 Phosphorylation Inhibits Fyn Kinase Interaction and Increases the Decay Time of NMDAR-mediated Current. J Mol Biol 2024; 436:168445. [PMID: 38218365 DOI: 10.1016/j.jmb.2024.168445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Fyn kinase SH3 domain interaction with PXXP motif in the Tau protein is implicated in AD pathology and is central to NMDAR function. Among seven PXXP motifs localized in proline-rich domain of Tau protein, tandem 5th and 6th PXXP motifs are critical to Fyn-SH3 domain interaction. Here, we report the crystal structure of Fyn-SH3 -Tau (207-221) peptide consisting of 5th and 6th PXXP motif complex to 1.01 Å resolution. Among five AD-specific phosphorylation sites encompassing the 5th and 6th PXXP motifs, only S214 residue showed interaction with SH3 domain. Biophysical studies showed that Tau (207-221) with S214-phosphorylation (pS214) inhibits its interaction with Fyn-SH3 domain. The individual administration of Tau (207-221) with/without pS214 peptides to a single neuron increased the decay time of evoked NMDA current response. Recordings of spontaneous NMDA EPSCs at +40 mV indicate an increase in frequency and amplitude of events for the Tau (207-221) peptide. Conversely, the Tau (207-221) with pS214 peptide exhibited a noteworthy amplitude increase alongside a prolonged decay time. These outcomes underscore the distinctive modalities of action associated with each peptide in the study. Overall, this study provides insights into how Tau (207-221) with/without pS214 affects the molecular framework of NMDAR signaling, indicating its involvement in Tau-related pathogenesis.
Collapse
Affiliation(s)
- Sneha Jos
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Roshni Poulose
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Archanalakshmi Kambaru
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Hemanga Gogoi
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | | | - Balasundaram Padmanabhan
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Bhupesh Mehta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| | - Sivaraman Padavattan
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| |
Collapse
|
3
|
Zhang H, Hu S, Yang P, Long H, Ma Q, Yin D, Xu G. HDAC9-mediated calmodulin deacetylation induces memory impairment in Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14573. [PMID: 38421101 PMCID: PMC10850929 DOI: 10.1111/cns.14573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/20/2023] [Accepted: 12/04/2023] [Indexed: 03/02/2024] Open
Abstract
AIMS Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive dysfunction and memory impairment. AD pathology involves protein acetylation. Previous studies have mainly focused on histone acetylation in AD, however, the roles of nonhistone acetylation in AD are less explored. METHODS The protein acetylation and expression levels were detected by western blotting and co-immunoprecipitation. The stoichiometry of acetylation was measured by home-made and site-specific antibodies against acetylated-CaM (Ac-CaM) at K22, K95, and K116. Hippocampus-dependent learning and memory were evaluated by using the Morris water maze, novel object recognition, and contextual fear conditioning tests. RESULTS We showed that calmodulin (CaM) acetylation is reduced in plasma of AD patients and mice. CaM acetylation and its target Ca2+ /CaM-dependent kinase II α (CaMKIIα) activity were severely impaired in AD mouse brain. The stoichiometry showed that Ac-K22, K95-CaM acetylation were decreased in AD patients and mice. Moreover, we screened and identified that lysine deacetylase 9 (HDAC9) was the main deacetylase for CaM. In addition, HDAC9 inhibition increased CaM acetylation and CaMKIIα activity, and hippocampus-dependent memory in AD mice. CONCLUSIONS HDAC9-mediated CaM deacetylation induces memory impairment in AD, HDAC9, or CaM acetylation may become potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Hai‐Long Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSuzhou Medical College of Soochow University, Medical Center of Soochow UniversitySuzhouChina
| | - Shufen Hu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSuzhou Medical College of Soochow University, Medical Center of Soochow UniversitySuzhouChina
| | - Pin Yang
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life ScienceEast China Normal UniversityShanghaiChina
| | - Han‐Chun Long
- Department of NeurologyThe Affiliated Xingyi City Hospital of Guizhou Medical UniversityXingyiChina
| | - Quan‐Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSuzhou Medical College of Soochow University, Medical Center of Soochow UniversitySuzhouChina
| | - Dong‐Min Yin
- Key Laboratory of Brain Functional Genomics, Ministry of Education and Shanghai, School of Life ScienceEast China Normal UniversityShanghaiChina
| | - Guang‐Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of NeuroscienceSuzhou Medical College of Soochow University, Medical Center of Soochow UniversitySuzhouChina
| |
Collapse
|
4
|
Hidalgo C, Paula-Lima A. RyR-mediated calcium release in hippocampal health and disease. Trends Mol Med 2024; 30:25-36. [PMID: 37957056 DOI: 10.1016/j.molmed.2023.10.008] [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: 09/07/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023]
Abstract
Hippocampal synaptic plasticity is widely considered the cellular basis of learning and spatial memory processes. This article highlights the central role of Ca2+ release from the endoplasmic reticulum (ER) in hippocampal synaptic plasticity and hippocampus-dependent memory in health and disease. The key participation of ryanodine receptor (RyR) channels, which are the principal Ca2+ release channels expressed in the hippocampus, in these processes is emphasized. It is proposed that the increased neuronal oxidative tone displayed by hippocampal neurons during aging or Alzheimer's disease (AD) leads to excessive activation of RyR-mediated Ca2+ release, a process that is highly redox-sensitive, and that this abnormal response contributes to and aggravates these deleterious conditions.
Collapse
Affiliation(s)
- Cecilia Hidalgo
- Biomedical Neuroscience Institute and Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; Physiology and Biophysics Program, Institute of Biomedical Sciences and Center for Exercise, Metabolism, and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile.
| | - Andrea Paula-Lima
- Biomedical Neuroscience Institute and Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; Institute for Research in Dental Sciences (ICOD), Faculty of Dentistry, Universidad de Chile, Santiago 8380544, Chile.
| |
Collapse
|
5
|
de Wet S, Theart R, Loos B. Cogs in the autophagic machine-equipped to combat dementia-prone neurodegenerative diseases. Front Mol Neurosci 2023; 16:1225227. [PMID: 37720551 PMCID: PMC10500130 DOI: 10.3389/fnmol.2023.1225227] [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: 05/18/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023] Open
Abstract
Neurodegenerative diseases are often characterized by hydrophobic inclusion bodies, and it may be the case that the aggregate-prone proteins that comprise these inclusion bodies are in fact the cause of neurotoxicity. Indeed, the appearance of protein aggregates leads to a proteostatic imbalance that causes various interruptions in physiological cellular processes, including lysosomal and mitochondrial dysfunction, as well as break down in calcium homeostasis. Oftentimes the approach to counteract proteotoxicity is taken to merely upregulate autophagy, measured by an increase in autophagosomes, without a deeper assessment of contributors toward effective turnover through autophagy. There are various ways in which autophagy is regulated ranging from the mammalian target of rapamycin (mTOR) to acetylation status of proteins. Healthy mitochondria and the intracellular energetic charge they preserve are key for the acidification status of lysosomes and thus ensuring effective clearance of components through the autophagy pathway. Both mitochondria and lysosomes have been shown to bear functional protein complexes that aid in the regulation of autophagy. Indeed, it may be the case that minimizing the proteins associated with the respective neurodegenerative pathology may be of greater importance than addressing molecularly their resulting inclusion bodies. It is in this context that this review will dissect the autophagy signaling pathway, its control and the manner in which it is molecularly and functionally connected with the mitochondrial and lysosomal system, as well as provide a summary of the role of autophagy dysfunction in driving neurodegenerative disease as a means to better position the potential of rapamycin-mediated bioactivities to control autophagy favorably.
Collapse
Affiliation(s)
- Sholto de Wet
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Rensu Theart
- Department of Electric and Electronic Engineering, Stellenbosch University, Stellenbosch, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
6
|
Nageeb Hasan SM, Clarke CL, McManamon Strand TP, Bambico FR. Putative pathological mechanisms of late-life depression and Alzheimer's Disease. Brain Res 2023:148423. [PMID: 37244602 DOI: 10.1016/j.brainres.2023.148423] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by progressive impairment in cognition and memory. AD is accompanied by several neuropsychiatric symptoms, with depression being the most prominent. Although depression has long been known to be associated with AD, controversial findings from preclinical and clinical studies have obscured the precise nature of this association. However recent evidence suggests that depression could be a prodrome or harbinger of AD. Evidence indicates that the major central serotonergic nucleus-the dorsal raphe nucleus (DRN)-shows very early AD pathology: neurofibrillary tangles made of hyperphosphorylated tau protein and degenerated neurites. AD and depression share common pathophysiologies, including functional deficits of the serotonin (5-HT) system. 5-HT receptors have modulatory effects on the progression of AD pathology i.e., reduction in Aβ load, increased hyper-phosphorylation of tau, decreased oxidative stress etc. Moreover, preclinical models show a role for specific channelopathies that result in abnormal regional activational and neuroplasticity patterns. One of these concerns the pathological upregulation of the small conductance calcium-activated potassium (SK) channel in corticolimbic structure. This has also been observed in the DRN in both diseases. The SKC is a key regulator of cell excitability and long-term potentiation (LTP). SKC over-expression is positively correlated with aging and cognitive decline, and is evident in AD. Pharmacological blockade of SKCs has been reported to reverse symptoms of depression and AD. Thus, aberrant SKC functioning could be related to depression pathophysiology and diverts its late-life progression towards the development of AD. We summarize findings from preclinical and clinical studies suggesting a molecular linkage between depression and AD pathology. We also provide a rationale for considering SKCs as a novel pharmacological target for the treatment of AD-associated symptoms.
Collapse
Affiliation(s)
- S M Nageeb Hasan
- Department of Psychology, Memorial University of Newfoundland and Labrador, Newfoundland and Labrador, A1B3Xs, Canada.
| | - Courtney Leigh Clarke
- Department of Psychology, Memorial University of Newfoundland and Labrador, Newfoundland and Labrador, A1B3Xs, Canada
| | | | - Francis Rodriguez Bambico
- Department of Psychology, Memorial University of Newfoundland and Labrador, Newfoundland and Labrador, A1B3Xs, Canada; Behavioural Neurobiology Laboratory, Centre for Addiction and Mental Health, Toronto, ON, M5T1R8, Canada
| |
Collapse
|
7
|
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: 16] [Impact Index Per Article: 16.0] [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
|
8
|
Hyperoside alleviates toxicity of β-amyloid via endoplasmic reticulum-mitochondrial calcium signal transduction cascade in APP/PS1 double transgenic Alzheimer's disease mice. Redox Biol 2023; 61:102637. [PMID: 36821955 PMCID: PMC9975698 DOI: 10.1016/j.redox.2023.102637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Alzheimer's disease is a neurodegenerative disorder characterized by a decline in cognitive function. The β-amyloid (Aβ) hypothesis suggests that Aβ peptides can spontaneously aggregate into β-fragment-containing oligomers and protofibrils, and this activation of the amyloid pathway alters Ca2+ signaling in neurons, leading to neurotoxicity and thus apoptosis of neuronal cells. In our study, a blood-brain barrier crossing flavonol glycoside hyperoside was identified with anti-Aβ aggregation, BACE inhibitory, and neuroprotective effect in cellular or APP/PSEN1 double transgenic Alzheimer's disease mice model. While our pharmacokinetic data confirmed that intranasal administration of hyperoside resulted in a higher bio-availability in mice brain, further in vivo studies revealed that it improved motor deficit, spatial memory and learning ability of APP/PSEN1 mice with reducing level of Aβ plaques and GFAP in the cortex and hippocampus. Bioinformatics, computational docking and in vitro assay results suggested that hyperoside bind to Aβ and interacted with ryanodine receptors, then regulated cellular apoptosis via endoplasmic reticulum-mitochondrial calcium (Ca2+) signaling pathway. Consistently, it was confirmed that hyperoside increased Bcl2, decreased Bax and cyto-c protein levels, and ameliorated neuronal cell death in both in vitro and in vivo model. By regulating Aβ-induced cell death via regulation on Ca2+ signaling cascade and mitochondrial membrane potential, our study suggested that hyperoside may work as a potential therapeutic agent or preventive remedy for Alzheimer's disease.
Collapse
|
9
|
Wiseman AL, Briggs CA, Peritt A, Kapecki N, Peterson DA, Shim SS, Stutzmann GE. Lithium Provides Broad Therapeutic Benefits in an Alzheimer's Disease Mouse Model. J Alzheimers Dis 2023; 91:273-290. [PMID: 36442195 DOI: 10.3233/jad-220758] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disorder with a progressive loss of cognitive function. Currently, no effective treatment regimen is available. Lithium, a mood stabilizer for bipolar disorder, exerts broad neuroprotective and neurotrophic actions and improves cognitive function. OBJECTIVE The study investigated if lithium stabilizes Ca2+ signaling abnormalities in hippocampal neurons and subsequently normalize downstream effects on AD neuropathology and synaptic plasticity in young AD mice. METHODS Four-month-old 3xTg-AD mice were treated with a LiCl diet chow for 30 days. At the end of the lithium treatment, a combination of two-photon Ca2+ imaging, electrophysiology, and immunohistochemistry assays were used to assess the effects of the LiCl treatment on inositol trisphosphate receptor (IP3R)-dependent endoplasmic reticulum (ER) Ca2+ and voltage-gated Ca2+ channel (VGCC)-mediated Ca2+ signaling in CA1 neurons, neuronal nitric oxide synthase (nNOS) and hyperphosphorylated tau (p-tau) levels and synaptic plasticity in the hippocampus and overlying cortex from 3xTg-ADmice. RESULTS Thirty-day LiCl treatment reduced aberrant IP3R-dependent ER Ca2+ and VGCC-mediated Ca2+ signaling in CA1 pyramidal neurons from 3xTg-AD mice and restored neuronal nitric oxide synthase (nNOS) and hyperphosphorylated tau (p-tau) levels to control levels in the hippocampal subfields and overlying cortex. The LiCl treatment enhanced post-tetanic potentiation (PTP), a form of short-term plasticity in the hippocampus. CONCLUSION The study found that lithium exerts therapeutic effects across several AD-associated early neuronal signaling abnormalities including aberrant Ca2+ signaling, nNOS, and p-tau formation and enhances short-term synaptic plasticity. Lithium could serve as an effective treatment or co-therapeutic for AD.
Collapse
Affiliation(s)
- Alyssa L Wiseman
- Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA.,School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL, USA
| | - Clark A Briggs
- Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA
| | - Ariel Peritt
- Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA.,Sackler School of Medicine, New York State/American Program of Tel Aviv University, Tel Aviv, Israel
| | - Nicolas Kapecki
- Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA
| | - Daniel A Peterson
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University, North Chicago, IL, USA.,Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA.,School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL, USA
| | - Seong S Shim
- Discipline of Psychiatry and Behavioral Sciences, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA.,Captain James A. Lovell Federal Health Care Center, Mental Health, North Chicago, IL, USA
| | - Grace E Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University, North Chicago, IL, USA.,Discipline of Neuroscience, The Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA.,School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, IL, USA
| |
Collapse
|
10
|
Mustaly-Kalimi S, Gallegos W, Marr RA, Gilman-Sachs A, Peterson DA, Sekler I, Stutzmann GE. Protein mishandling and impaired lysosomal proteolysis generated through calcium dysregulation in Alzheimer's disease. Proc Natl Acad Sci U S A 2022; 119:e2211999119. [PMID: 36442130 PMCID: PMC9894236 DOI: 10.1073/pnas.2211999119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022] Open
Abstract
Impairments in neural lysosomal- and autophagic-mediated degradation of cellular debris contribute to neuritic dystrophy and synaptic loss. While these are well-characterized features of neurodegenerative disorders such as Alzheimer's disease (AD), the upstream cellular processes driving deficits in pathogenic protein mishandling are less understood. Using a series of fluorescent biosensors and optical imaging in model cells, AD mouse models and human neurons derived from AD patients, we reveal a previously undescribed cellular signaling cascade underlying protein mishandling mediated by intracellular calcium dysregulation, an early component of AD pathogenesis. Increased Ca2+ release via the endoplasmic reticulum (ER)-resident ryanodine receptor (RyR) is associated with reduced expression of the lysosome proton pump vacuolar-ATPase (vATPase) subunits (V1B2 and V0a1), resulting in lysosome deacidification and disrupted proteolytic activity in AD mouse models and human-induced neurons (HiN). As a result of impaired lysosome digestive capacity, mature autophagosomes with hyperphosphorylated tau accumulated in AD murine neurons and AD HiN, exacerbating proteinopathy. Normalizing AD-associated aberrant RyR-Ca2+ signaling with the negative allosteric modulator, dantrolene (Ryanodex), restored vATPase levels, lysosomal acidification and proteolytic activity, and autophagic clearance of intracellular protein aggregates in AD neurons. These results highlight that prior to overt AD histopathology or cognitive deficits, aberrant upstream Ca2+ signaling disrupts lysosomal acidification and contributes to pathological accumulation of intracellular protein aggregates. Importantly, this is demonstrated in animal models of AD, and in human iPSC-derived neurons from AD patients. Furthermore, pharmacological suppression of RyR-Ca2+ release rescued proteolytic function, revealing a target for therapeutic intervention that has demonstrated effects in clinically-relevant assays.
Collapse
Affiliation(s)
- Sarah Mustaly-Kalimi
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Wacey Gallegos
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Robert A. Marr
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Alice Gilman-Sachs
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University of Medicine and Science, Immunology, and Infection, North Chicago, IL60064
| | - Daniel A. Peterson
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Science and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva84105, Israel
| | - Grace E. Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| |
Collapse
|
11
|
Tozlu ÖÖ, Türkez H, Okkay U, Ceylan O, Bayram C, Hacımüftüoğlu A, Mardinoğlu A. Assessment of the neuroprotective potential of d-cycloserine and l-serine in aluminum chloride-induced experimental models of Alzheimer’s disease: In vivo and in vitro studies. Front Nutr 2022; 9:981889. [PMID: 36159454 PMCID: PMC9493202 DOI: 10.3389/fnut.2022.981889] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease characterized by the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles in the brain accompanied by synaptic dysfunction and neurodegeneration. No effective treatment has been found to slow the progression of the disease. Therapeutic studies using experimental animal models have therefore become very important. Therefore, this study aimed to investigate the possible neuroprotective effect of D-cycloserine and L-serine against aluminum chloride (AlCl3)-induced AD in rats. Administration of AlCl3 for 28 days caused oxidative stress and neurodegeneration compared to the control group. In addition, we found that aluminum decreases α-secretase activity while increasing β-secretase and γ-secretase activities by molecular genetic analysis. D-cycloserine and L-serine application resulted in an improvement in neurodegeneration and oxidative damage caused by aluminum toxicity. It is believed that the results of this study will contribute to the synthesis of new compounds with improved potential against AlCl3-induced neurodegeneration, cognitive impairment, and drug development research.
Collapse
Affiliation(s)
- Özlem Özdemir Tozlu
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, Turkey
| | - Hasan Türkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Ufuk Okkay
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Onur Ceylan
- Department of Medical Pathology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Cemil Bayram
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Ahmet Hacımüftüoğlu
- Department of Medical Pharmacology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Adil Mardinoğlu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, United Kingdom
- *Correspondence: Adil Mardinoğlu,
| |
Collapse
|
12
|
Abstract
Pannexin-1 (Panx1) channels contribute to neurological disorders, including stroke and epilepsy, where their function has been linked to N-methyl D-aspartate (NMDA) receptors (NMDARs). We discovered that Ca2+ entry via NMDARs recruits endoplasmic reticulum–resident STIM proteins to activate Panx1 by binding to a hydrophobic region localized to the Panx1 N terminus. Using loss-of-function approaches, combined with molecular replacement and use of a STIM/Panx1 function–blocking antibody, we demonstrate that disrupting the STIM/Panx1 interaction prevents Panx1 activation by NMDARs, but not by hypotonic stimuli. Thus, our findings serve as a basis for the design of modality-specific inhibitors against STIM-dependent Panx1 activation that will aid in understanding the multimodal functions of Panx1 and their contribution to physiology and pathology. Pannexin-1 (Panx1) is a large-pore ion and solute permeable channel highly expressed in the nervous system, where it subserves diverse processes, including neurite outgrowth, dendritic spine formation, and N-methyl D-aspartate (NMDA) receptor (NMDAR)-dependent plasticity. Moreover, Panx1 dysregulation contributes to neurological disorders, including neuropathic pain, epilepsy, and excitotoxicity. Despite progress in understanding physiological and pathological functions of Panx1, the mechanisms that regulate its activity, including its ion and solute permeability, remain poorly understood. In this study, we identify endoplasmic reticulum (ER)-resident stromal interaction molecules (STIM1/2), which are Ca2+ sensors that communicate events within the ER to plasma membrane channels, as binding and signaling partners of Panx1. We demonstrate that Panx1 is activated to its large-pore configuration in response to stimuli that recruit STIM1/2 and map the interaction interface to a hydrophobic region within the N terminus of Panx1. We further characterize a Panx1 N terminus–recognizing antibody as a function-blocking tool able to prevent large-pore Panx1 activation by STIM1/2. Using either the function-blocking antibody or re-expression of Panx1 deletion mutants in Panx1 knockout (KO) neurons, we show that STIM recruitment couples Ca2+ entry via NMDARs to Panx1 activation, thereby identifying a model of NMDAR-STIM-Panx1 signaling in neurons. Our study highlights a previously unrecognized and important role of the Panx1 N terminus in regulating channel activation and membrane localization. Considering past work demonstrating an intimate functional relation between NMDARs and Panx1, our study opens avenues for understanding activation modality and context-specific functions of Panx1, including functions linked to diverse STIM-regulated cellular responses.
Collapse
|
13
|
Bouron A. Store-operated ion channels: a growing family ? Cell Calcium 2022; 107:102657. [DOI: 10.1016/j.ceca.2022.102657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/02/2022]
|
14
|
WEI HF, ANCHIPOLOVSKY S, VERA R, LIANG G, CHUANG DM. Potential mechanisms underlying lithium treatment for Alzheimer's disease and COVID-19. EUROPEAN REVIEW FOR MEDICAL AND PHARMACOLOGICAL SCIENCES 2022; 26:2201-2214. [PMID: 35363371 PMCID: PMC9173589 DOI: 10.26355/eurrev_202203_28369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Disruption of intracellular Ca2+ homeostasis plays an important role as an upstream pathology in Alzheimer's disease (AD), and correction of Ca2+ dysregulation has been increasingly proposed as a target of future effective disease-modified drugs for treating AD. Calcium dysregulation is also an upstream pathology for the COVID-19 virus SARS-CoV-2 infection and replication, leading to host cell damage. Clinically available drugs that can inhibit the disturbed intracellular Ca2+ homeostasis have been repurposed to treat COVID-19 patients. This narrative review aims at exploring the underlying mechanism by which lithium, a first line drug for the treatment of bipolar disorder, inhibits Ca2+ dysregulation and associated downstream pathology in both AD and COVID-19. It is suggested that lithium can be repurposed to treat AD patients, especially those afflicted with COVID-19.
Collapse
Affiliation(s)
- H.-F. WEI
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - S. ANCHIPOLOVSKY
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - R. VERA
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - G. LIANG
- Department of Anaesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - D.-M. CHUANG
- Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD, USA
| |
Collapse
|
15
|
Schreiner TG, Popescu BO. Impact of Caffeine on Alzheimer’s Disease Pathogenesis—Protective or Risk Factor? Life (Basel) 2022; 12:life12030330. [PMID: 35330081 PMCID: PMC8952218 DOI: 10.3390/life12030330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), the most common dementia worldwide, remains without an effective treatment to this day despite intensive research conducted during the last decades. In this context, researchers have turned their attention towards the prevention of this pathology, focusing on early detection and better control of the most important risk factors, concomitantly with trying to find potentially protective factors that may delay the onset of AD. From the multitude of factors studied, coffee (especially its main component, caffeine) is a current interesting research topic, taking into consideration the contradictory results of recent years’ studies. On the one hand, much of the evidence from fundamental research suggests the potentially protective trait of caffeine in AD, while other data mainly from human studies lean toward no correlation or even suggesting that caffeine is a veritable risk factor for dementia. Given the methodological heterogeneity of the studies, this review aims to bring new evidence regarding this topic and to try to clearly establish a correlation between the two entities. Thus, in the first part, the authors make a clear distinction between the effects of coffee and the effects of caffeine in AD, presenting a rich basis of clinical trials on both animal models and the human subject. Subsequently, the main pathophysiological mechanisms that would explain the action of caffeine in the etiopathogenesis of AD are reviewed. Finally, the role of computational models is presented, having beneficial impact on both better understanding of the disease mechanism and the development of new therapeutic approaches for AD prevention.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania;
- Department of Neurology, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
- Correspondence:
| | - Bogdan Ovidiu Popescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania;
- Neurology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
16
|
Paasila PJ, Aramideh JA, Sutherland GT, Graeber MB. Synapses, Microglia, and Lipids in Alzheimer's Disease. Front Neurosci 2022; 15:778822. [PMID: 35095394 PMCID: PMC8789683 DOI: 10.3389/fnins.2021.778822] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is characterised by synaptic dysfunction accompanied by the microscopically visible accumulation of pathological protein deposits and cellular dystrophy involving both neurons and glia. Late-stage AD shows pronounced loss of synapses and neurons across several differentially affected brain regions. Recent studies of advanced AD using post-mortem brain samples have demonstrated the direct involvement of microglia in synaptic changes. Variants of the Apolipoprotein E and Triggering Receptors Expressed on Myeloid Cells gene represent important determinants of microglial activity but also of lipid metabolism in cells of the central nervous system. Here we review evidence that may help to explain how abnormal lipid metabolism, microglial activation, and synaptic pathophysiology are inter-related in AD.
Collapse
Affiliation(s)
- Patrick J. Paasila
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Jason A. Aramideh
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Greg T. Sutherland
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Manuel B. Graeber
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
17
|
Liu L, Gao H, Zaikin A, Chen S. Unraveling Aβ-Mediated Multi-Pathway Calcium Dynamics in Astrocytes: Implications for Alzheimer's Disease Treatment From Simulations. Front Physiol 2021; 12:767892. [PMID: 34777023 PMCID: PMC8581622 DOI: 10.3389/fphys.2021.767892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/08/2021] [Indexed: 02/02/2023] Open
Abstract
The accumulation of amyloid β peptide (Aβ) in the brain is hypothesized to be the major factor driving Alzheimer's disease (AD) pathogenesis. Mounting evidence suggests that astrocytes are the primary target of Aβ neurotoxicity. Aβ is known to interfere with multiple calcium fluxes, thus disrupting the calcium homeostasis regulation of astrocytes, which are likely to produce calcium oscillations. Ca2+ dyshomeostasis has been observed to precede the appearance of clinical symptoms of AD; however, it is experimentally very difficult to investigate the interactions of many mechanisms. Given that Ca2+ disruption is ubiquitously involved in AD progression, it is likely that focusing on Ca2+ dysregulation may serve as a potential therapeutic approach to preventing or treating AD, while current hypotheses concerning AD have so far failed to yield curable therapies. For this purpose, we derive and investigate a concise mathematical model for Aβ-mediated multi-pathway astrocytic intracellular Ca2+ dynamics. This model accounts for how Aβ affects various fluxes contributions through voltage-gated calcium channels, Aβ-formed channels and ryanodine receptors. Bifurcation analysis of Aβ level, which reflected the corresponding progression of the disease, revealed that Aβ significantly induced the increasing [Ca2+] i and frequency of calcium oscillations. The influence of inositol 1,4,5-trisphosphate production (IP3) is also investigated in the presence of Aβ as well as the impact of changes in resting membrane potential. In turn, the Ca2+ flux can be considerably changed by exerting specific interventions, such as ion channel blockers or receptor antagonists. By doing so, a "combination therapy" targeting multiple pathways simultaneously has finally been demonstrated to be more effective. This study helps to better understand the effect of Aβ, and our findings provide new insight into the treatment of AD.
Collapse
Affiliation(s)
- Langzhou Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Huayi Gao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Alexey Zaikin
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.,Institute for Women's Health and Department of Mathematics, University College London, London, United Kingdom.,World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Shangbin Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Sanderson JL, Freund RK, Gorski JA, Dell'Acqua ML. β-Amyloid disruption of LTP/LTD balance is mediated by AKAP150-anchored PKA and Calcineurin regulation of Ca 2+-permeable AMPA receptors. Cell Rep 2021; 37:109786. [PMID: 34610314 PMCID: PMC8530450 DOI: 10.1016/j.celrep.2021.109786] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/02/2021] [Accepted: 09/10/2021] [Indexed: 01/28/2023] Open
Abstract
Regulated insertion and removal of postsynaptic AMPA glutamate receptors (AMPARs) mediates hippocampal long-term potentiation (LTP) and long-term depression (LTD) synaptic plasticity underlying learning and memory. In Alzheimer’s disease β-amyloid (Aβ) oligomers may impair learning and memory by altering AMPAR trafficking and LTP/LTD balance. Importantly, Ca2+-permeable AMPARs (CP-AMPARs) assembled from GluA1 subunits are excluded from hippocampal synapses basally but can be recruited rapidly during LTP and LTD to modify synaptic strength and signaling. By employing mouse knockin mutations that disrupt anchoring of the kinase PKA or phosphatase Calcineurin (CaN) to the postsynaptic scaffold protein AKAP150, we find that local AKAP-PKA signaling is required for CP-AMPAR recruitment, which can facilitate LTP but also, paradoxically, prime synapses for Aβ impairment of LTP mediated by local AKAP-CaN LTD signaling that promotes subsequent CP-AMPAR removal. These findings highlight the importance of PKA/CaN signaling balance and CP-AMPARs in normal plasticity and aberrant plasticity linked to disease. In Alzheimer’s disease, Aβ oligomers disrupt hippocampal neuronal plasticity and cognition. Sanderson et al. show how the postsynaptic scaffold protein AKAP150 coordinates PKA and Calcineurin regulation of Ca2+-permeable AMPA-type glutamate receptors to mediate disruption of synaptic plasticity by Aβ oligomers.
Collapse
Affiliation(s)
- Jennifer L Sanderson
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ronald K Freund
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jessica A Gorski
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA; University of Colorado Alzheimer's and Cognition Center, Anschutz Medical Campus, Aurora, CO 80045, USA; Linda Crnic Institute for Down Syndrome, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
19
|
Chiang TI, Yu YH, Lin CH, Lane HY. Novel Biomarkers of Alzheimer's Disease: Based Upon N-methyl-D-aspartate Receptor Hypoactivation and Oxidative Stress. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2021; 19:423-433. [PMID: 34294612 PMCID: PMC8316669 DOI: 10.9758/cpn.2021.19.3.423] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/07/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Early detection and prevention of Alzheimer’s disease (AD) is important. The current treatment for early AD is acetylcholine esterase inhibitors (AChEIs); however, the efficacy is poor. Besides, AChEI did not show efficacy in mild cognitive impairment (MCI). Beta-amyloid (Aβ) deposits have been regarded to be highly related to the pathogenesis of AD. However, many clinical trials aiming at the clearance of Aβ deposits failed to improve the cognitive decline of AD, even at its early phase. There should be other important mechanisms unproven in the course of AD and MCI. Feasible biomarkers for the diagnosis and treatment response of AD are lacking to date. The N-methyl-D-aspartate receptor (NMDAR) activation plays an important role in learning and memory. On the other hand, oxidative stress has been regarded to contribute to aging with the assumption that free radicals damage cell constituents and connective tissues. Our recent study found that an NMDAR enhancer, sodium benzoate (the pivotal inhibitor of D-amino acid oxidase [DAAO]), improved the cognitive and global function of patients with early-phase AD. Further, we found that peripheral DAAO levels were higher in patients with MCI and AD than healthy controls. We also found that sodium benzoate was able to change the activity of antioxidant. These pieces of evidence suggest that the NMDAR function is associated with anti-oxidation, and have potential to be biomarkers for the diagnosis and treatment response of AD.
Collapse
Affiliation(s)
- Ting-I Chiang
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Hsiang Yu
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chieh-Hsin Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Psychiatry and Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan.,Department of Psychology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
20
|
Chami M, Checler F. Targeting Post-Translational Remodeling of Ryanodine Receptor: A New Track for Alzheimer's Disease Therapy? Curr Alzheimer Res 2021; 17:313-323. [PMID: 32096743 DOI: 10.2174/1567205017666200225102941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/08/2020] [Accepted: 02/24/2020] [Indexed: 01/20/2023]
Abstract
Pathologic calcium (Ca2+) signaling linked to Alzheimer's Disease (AD) involves the intracellular Ca2+ release channels/ryanodine receptors (RyRs). RyRs are macromolecular complexes where the protein-protein interactions between RyRs and several regulatory proteins impact the channel function. Pharmacological and genetic approaches link the destabilization of RyRs macromolecular complexes to several human pathologies including brain disorders. In this review, we discuss our recent data, which demonstrated that enhanced neuronal RyR2-mediated Ca2+ leak in AD is associated with posttranslational modifications (hyperphosphorylation, oxidation, and nitrosylation) leading to RyR2 macromolecular complex remodeling, and dissociation of the stabilizing protein Calstabin2 from the channel. We describe RyR macromolecular complex structure and discuss the molecular mechanisms and signaling cascade underlying neuronal RyR2 remodeling in AD. We provide evidence linking RyR2 dysfunction with β-adrenergic signaling cascade that is altered in AD. RyR2 remodeling in AD leads to histopathological lesions, alteration of synaptic plasticity, learning and memory deficits. Targeting RyR macromolecular complex remodeling should be considered as a new therapeutic window to treat/or prevent AD setting and/or progression.
Collapse
Affiliation(s)
- Mounia Chami
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, F-06560, France.,CNRS, IPMC, Sophia Antipolis, F-06560, France
| | - Frédéric Checler
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, F-06560, France.,CNRS, IPMC, Sophia Antipolis, F-06560, France
| |
Collapse
|
21
|
Nagu P, Parashar A, Behl T, Mehta V. Gut Microbiota Composition and Epigenetic Molecular Changes Connected to the Pathogenesis of Alzheimer's Disease. J Mol Neurosci 2021; 71:1436-1455. [PMID: 33829390 DOI: 10.1007/s12031-021-01829-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, and its pathogenesis is not fully known. Although there are several hypotheses, such as neuroinflammation, tau hyperphosphorylation, amyloid-β plaques, neurofibrillary tangles, and oxidative stress, none of them completely explain the origin and progression of AD. Emerging evidence suggests that gut microbiota and epigenetics can directly influence the pathogenesis of AD via their effects on multiple pathways, including neuroinflammation, oxidative stress, and amyloid protein. Various gut microbes such as Actinobacteria, Bacteroidetes, E. coli, Firmicutes, Proteobacteria, Tenericutes, and Verrucomicrobia are known to play a crucial role in the pathogenesis of AD. These microbes and their metabolites modulate various physiological processes that contribute to AD pathogenesis, such as neuroinflammation and other inflammatory processes, amyloid deposition, cytokine storm syndrome, altered BDNF and NMDA signaling, impairing neurodevelopmental processes. Likewise, epigenetic markers associated with AD mainly include histone modifications and DNA methylation, which are under the direct control of a variety of enzymes, such as acetylases and methylases. The activity of these enzymes is dependent upon the metabolites generated by the host's gut microbiome, suggesting the significance of epigenetics in AD pathogenesis. It is interesting to know that both gut microbiota and epigenetics are dynamic processes and show a high degree of variation according to diet, stressors, and environmental factors. The bidirectional relation between the gut microbiota and epigenetics suggests that they might work in synchrony to modulate AD representation, its pathogenesis, and progression. They both also provide numerous targets for early diagnostic biomarkers and for the development of AD therapeutics. This review discusses the gut microbiota and epigenetics connection in the pathogenesis of AD and aims to highlight vast opportunities for diagnosis and therapeutics of AD.
Collapse
Affiliation(s)
- Priyanka Nagu
- Department of Pharmaceutics, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.,Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala University, Jhunjhunu, Rajasthan, India
| | - Arun Parashar
- Faculty of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Department of Pharmacology, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.
| |
Collapse
|
22
|
Potential of Caffeine in Alzheimer's Disease-A Review of Experimental Studies. Nutrients 2021; 13:nu13020537. [PMID: 33562156 PMCID: PMC7915779 DOI: 10.3390/nu13020537] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia leading to progressive memory loss and cognitive impairment. Considering that pharmacological treatment options for AD are few and not satisfactory, increasing attention is being paid to dietary components that may affect the development of the disease. Such a dietary component may be caffeine contained in coffee, tea or energy drinks. Although epidemiological data suggest that caffeine intake may counteract the development of cognitive impairment, results of those studies are not conclusive. The aim of the present study is to review the existing experimental studies on the efficacy of caffeine against AD and AD-related cognitive impairment, focusing on the proposed protective mechanisms of action. In conclusion, the reports of studies on experimental AD models generally supported the notion that caffeine may exert some beneficial effects in AD. However, further studies are necessary to elucidate the role of caffeine in the effects of its sources on cognition and possibly AD risk.
Collapse
|
23
|
Ca 2+ Dyshomeostasis Disrupts Neuronal and Synaptic Function in Alzheimer's Disease. Cells 2020; 9:cells9122655. [PMID: 33321866 PMCID: PMC7763805 DOI: 10.3390/cells9122655] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Ca2+ homeostasis is essential for multiple neuronal functions and thus, Ca2+ dyshomeostasis can lead to widespread impairment of cellular and synaptic signaling, subsequently contributing to dementia and Alzheimer's disease (AD). While numerous studies implicate Ca2+ mishandling in AD, the cellular basis for loss of cognitive function remains under investigation. The process of synaptic degradation and degeneration in AD is slow, and constitutes a series of maladaptive processes each contributing to a further destabilization of the Ca2+ homeostatic machinery. Ca2+ homeostasis involves precise maintenance of cytosolic Ca2+ levels, despite extracellular influx via multiple synaptic Ca2+ channels, and intracellular release via organelles such as the endoplasmic reticulum (ER) via ryanodine receptor (RyRs) and IP3R, lysosomes via transient receptor potential mucolipin channel (TRPML) and two pore channel (TPC), and mitochondria via the permeability transition pore (PTP). Furthermore, functioning of these organelles relies upon regulated inter-organelle Ca2+ handling, with aberrant signaling resulting in synaptic dysfunction, protein mishandling, oxidative stress and defective bioenergetics, among other consequences consistent with AD. With few effective treatments currently available to mitigate AD, the past few years have seen a significant increase in the study of synaptic and cellular mechanisms as drivers of AD, including Ca2+ dyshomeostasis. Here, we detail some key findings and discuss implications for future AD treatments.
Collapse
|
24
|
Wang Y, Shi Z, Zhang Y, Yan J, Yu W, Chen L. Oligomer β-amyloid Induces Hyperactivation of Ras to Impede NMDA Receptor-Dependent Long-Term Potentiation in Hippocampal CA1 of Mice. Front Pharmacol 2020; 11:595360. [PMID: 33536910 PMCID: PMC7848859 DOI: 10.3389/fphar.2020.595360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/20/2020] [Indexed: 12/02/2022] Open
Abstract
The activity of Ras, a small GTPase protein, is increased in brains with Alzheimer’s disease. The objective of this study was to determine the influence of oligomeric Aβ1-42 on the activation of Ras, and the involvement of the Ras hyperactivity in Aβ1-42-induced deficits in spatial cognition and hippocampal synaptic plasticity. Herein, we show that intracerebroventricular injection of Aβ1-42 in mice (Aβ-mice) enhanced hippocampal Ras activation and expression, while 60 min incubation of hippocampal slices in Aβ1-42 (Aβ-slices) only elevated Ras activity. Aβ-mice showed deficits in spatial cognition and NMDA receptor (NMDAR)-dependent long-term potentiation (LTP) in hippocampal CA1, but basal synaptic transmission was enhanced. The above effects of Aβ1-42 were corrected by the Ras inhibitor farnesylthiosalicylic acid (FTS). ERK2 phosphorylation increased, and Src phosphorylation decreased in Aβ-mice and Aβ1-42-slices. Both were corrected by FTS. In CA1 pyramidal cells of Aβ1-42-slices, the response of AMPA receptor and phosphorylation of GluR1 were enhanced with dependence on Ras activation rather than ERK signaling. In contrast, NMDA receptor (NMDAR) function and GluN2A/2B phosphorylation were downregulated in Aβ1-42-slices, which was recovered by application of FTS or the Src activator ouabain, and mimicked in control slices treated with the Src inhibitor PP2. The administration of PP2 impaired the spatial cognition and LTP induction in control mice and FTS-treated Aβ-mice. The treatment of Aβ-mice with ouabain rescued Aβ-impaired spatial cognition and LTP. Overall, the results indicate that the oligomeric Aβ1-42 hyperactivates Ras and thereby causes the downregulation of Src which impedes NMDAR-dependent LTP induction resulting in cognitive deficits.
Collapse
Affiliation(s)
- Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Zhaochun Shi
- Department of Neurology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yajie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Jun Yan
- Department of Geriatric Medicine, Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wenfeng Yu
- Key Laboratory of Endemic and Ethnic Diseases of Education Ministry, Guizhou Medical University, Guizhou, China
| | - Ling Chen
- Department of Physiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Chami M, Checler F. Alterations of the Endoplasmic Reticulum (ER) Calcium Signaling Molecular Components in Alzheimer's Disease. Cells 2020; 9:cells9122577. [PMID: 33271984 PMCID: PMC7760721 DOI: 10.3390/cells9122577] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/18/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Sustained imbalance in intracellular calcium (Ca2+) entry and clearance alters cellular integrity, ultimately leading to cellular homeostasis disequilibrium and cell death. Alzheimer’s disease (AD) is the most common cause of dementia. Beside the major pathological features associated with AD-linked toxic amyloid beta (Aβ) and hyperphosphorylated tau (p-tau), several studies suggested the contribution of altered Ca2+ handling in AD development. These studies documented physical or functional interactions of Aβ with several Ca2+ handling proteins located either at the plasma membrane or in intracellular organelles including the endoplasmic reticulum (ER), considered the major intracellular Ca2+ pool. In this review, we describe the cellular components of ER Ca2+ dysregulations likely responsible for AD. These include alterations of the inositol 1,4,5-trisphosphate receptors’ (IP3Rs) and ryanodine receptors’ (RyRs) expression and function, dysfunction of the sarco-endoplasmic reticulum Ca2+ ATPase (SERCA) activity and upregulation of its truncated isoform (S1T), as well as presenilin (PS1, PS2)-mediated ER Ca2+ leak/ER Ca2+ release potentiation. Finally, we highlight the functional consequences of alterations of these ER Ca2+ components in AD pathology and unravel the potential benefit of targeting ER Ca2+ homeostasis as a tool to alleviate AD pathogenesis.
Collapse
Affiliation(s)
- Mounia Chami
- Correspondence: ; Tel.: +33-4939-53457; Fax: +33-4939-53408
| | | |
Collapse
|
26
|
Ghoweri AO, Ouillette L, Frazier HN, Anderson KL, Lin RL, Gant JC, Parent R, Moore S, Murphy GG, Thibault O. Electrophysiological and Imaging Calcium Biomarkers of Aging in Male and Female 5×FAD Mice. J Alzheimers Dis 2020; 78:1419-1438. [PMID: 33164928 PMCID: PMC7836067 DOI: 10.3233/jad-200109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND In animal models and tissue preparations, calcium dyshomeostasis is a biomarker of aging and Alzheimer's disease that is associated with synaptic dysfunction, neuritic pruning, and dysregulated cellular processes. It is unclear, however, whether the onset of calcium dysregulation precedes, is concurrent with, or is the product of pathological cellular events (e.g., oxidation, amyloid-β production, and neuroinflammation). Further, neuronal calcium dysregulation is not always present in animal models of amyloidogenesis, questioning its reliability as a disease biomarker. OBJECTIVE Here, we directly tested for the presence of calcium dysregulation in dorsal hippocampal neurons in male and female 5×FAD mice on a C57BL/6 genetic background using sharp electrodes coupled with Oregon-green Bapta-1 imaging. We focused on three ages that coincide with the course of amyloid deposition: 1.5, 4, and 10 months old. METHODS Outcome variables included measures of the afterhyperpolarization, short-term synaptic plasticity, and calcium kinetics during synaptic activation. Quantitative analyses of spatial learning and memory were also conducted using the Morris water maze. Main effects of sex, age, and genotype were identified on measures of electrophysiology and calcium imaging. RESULTS Measures of resting Oregon-green Bapta-1 fluorescence showed significant reductions in the 5×FAD group compared to controls. Deficits in spatial memory, along with increases in Aβ load, were detectable at older ages, allowing us to test for temporal associations with the onset of calcium dysregulation. CONCLUSION Our results provide evidence that reduced, rather than elevated, neuronal calcium is identified in this 5×FAD model and suggests that this surprising result may be a novel biomarker of AD.
Collapse
Affiliation(s)
- Adam O Ghoweri
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Lara Ouillette
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Hilaree N Frazier
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Katie L Anderson
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ruei-Lung Lin
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - John C Gant
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Rachel Parent
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Shannon Moore
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,5037 BSRB, Molecular and Integrative Physiology, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Geoffrey G Murphy
- 5037 BSRB, Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,5037 BSRB, Molecular and Integrative Physiology, Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Olivier Thibault
- UKMC MS313, Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
27
|
Wang ZT, Zhang C, Wang YJ, Dong Q, Tan L, Yu JT. Selective neuronal vulnerability in Alzheimer's disease. Ageing Res Rev 2020; 62:101114. [PMID: 32569730 DOI: 10.1016/j.arr.2020.101114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is defined by a deficiency in specific behavioural and/or cognitive domains, pointing to selective vulnerabilities of specific neurons from different brain regions. These vulnerabilities can be compared across neuron subgroups to identify the most vulnerable neuronal types, regions, and time points for further investigation. Thus, the relevant organizational frameworks for brain subgroups will hold great values for a clear understanding of the progression in AD. Presently, the neuronal vulnerability has yet urgently required to be elucidated as not yet been clearly defined. It is suggested that cell-autonomous and non-cell-autonomous mechanisms can affect the neuronal vulnerability to stressors, and in turn modulates AD progression. This review examines cell-autonomous and non-cell-autonomous mechanisms that contribute to the neuronal vulnerability. Collectively, the cell-autonomous mechanisms seem to be the primary drivers responsible for initiating specific stressor-related neuronal vulnerability with pathological changes in certain brain areas, which then utilize non-cell-autonomous mechanisms and result in subsequent progression of AD. In summary, this article has provided a new perspective on the preventative and therapeutic options for AD.
Collapse
Affiliation(s)
- Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Diseases (MIND), Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129-2060, USA
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China; Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
28
|
Agrawal RR, Montesinos J, Larrea D, Area-Gomez E, Pera M. The silence of the fats: A MAM's story about Alzheimer. Neurobiol Dis 2020; 145:105062. [PMID: 32866617 DOI: 10.1016/j.nbd.2020.105062] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/07/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023] Open
Abstract
The discovery of contact sites was a breakthrough in cell biology. We have learned that an organelle cannot function in isolation, and that many cellular functions depend on communication between two or more organelles. One such contact site results from the close apposition of the endoplasmic reticulum (ER) and mitochondria, known as mitochondria-associated ER membranes (MAMs). These intracellular lipid rafts serve as hubs for the regulation of cellular lipid and calcium homeostasis, and a growing body of evidence indicates that MAM domains modulate cellular function in both health and disease. Indeed, MAM dysfunction has been described as a key event in Alzheimer disease (AD) pathogenesis. Our most recent work shows that, by means of its affinity for cholesterol, APP-C99 accumulates in MAM domains of the ER and induces the uptake of extracellular cholesterol as well as its trafficking from the plasma membrane to the ER. As a result, MAM functionality becomes chronically upregulated while undergoing continual turnover. The goal of this review is to discuss the consequences of C99 elevation in AD, specifically the upregulation of cholesterol trafficking and MAM activity, which abrogate cellular lipid homeostasis and disrupt the lipid composition of cellular membranes. Overall, we present a novel framework for AD pathogenesis that can be linked to the many complex alterations that occur during disease progression, and that may open a door to new therapeutic strategies.
Collapse
Affiliation(s)
- Rishi R Agrawal
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jorge Montesinos
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Delfina Larrea
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Estela Area-Gomez
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, 10032, USA; Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Marta Pera
- Departament of Basic Sciences, Facultat de Medicina I Ciències de la Salut, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallés, 08195, Spain.
| |
Collapse
|
29
|
Schrank S, Barrington N, Stutzmann GE. Calcium-Handling Defects and Neurodegenerative Disease. Cold Spring Harb Perspect Biol 2020; 12:a035212. [PMID: 31427373 PMCID: PMC7328457 DOI: 10.1101/cshperspect.a035212] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Calcium signaling is critical to neuronal function and regulates highly diverse processes such as gene transcription, energy production, protein handling, and synaptic structure and function. Because there are many common underlying calcium-mediated pathological features observed across several neurological conditions, it has been proposed that neurodegenerative diseases have an upstream underlying calcium basis in their pathogenesis. With certain diseases such as Alzheimer's, Parkinson's, and Huntington's, specific sources of calcium dysregulation originating from distinct neuronal compartments or channels have been shown to have defined roles in initiating or sustaining disease mechanisms. Herein, we will review the major hallmarks of these diseases, and how they relate to calcium dysregulation. We will then discuss neuronal calcium handling throughout the neuron, with special emphasis on channels involved in neurodegeneration.
Collapse
Affiliation(s)
- Sean Schrank
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University, North Chicago, Illinois 60064
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, Illinois 60064
| | - Nikki Barrington
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University, North Chicago, Illinois 60064
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, Illinois 60064
- Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois 60064
| | - Grace E Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University, North Chicago, Illinois 60064
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University, North Chicago, Illinois 60064
- Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois 60064
| |
Collapse
|
30
|
García F, Lobos P, Ponce A, Cataldo K, Meza D, Farías P, Estay C, Oyarzun-Ampuero F, Herrera-Molina R, Paula-Lima A, Ardiles ÁO, Hidalgo C, Adasme T, Muñoz P. Astaxanthin Counteracts Excitotoxicity and Reduces the Ensuing Increases in Calcium Levels and Mitochondrial Reactive Oxygen Species Generation. Mar Drugs 2020; 18:md18060335. [PMID: 32604880 PMCID: PMC7345213 DOI: 10.3390/md18060335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/28/2020] [Accepted: 06/08/2020] [Indexed: 12/17/2022] Open
Abstract
Astaxanthin (ASX) is a carotenoid pigment with strong antioxidant properties. We have reported previously that ASX protects neurons from the noxious effects of amyloid-β peptide oligomers, which promote excessive mitochondrial reactive oxygen species (mROS) production and induce a sustained increase in cytoplasmic Ca2+ concentration. These properties make ASX a promising therapeutic agent against pathological conditions that entail oxidative and Ca2+ dysregulation. Here, we studied whether ASX protects neurons from N-methyl-D-aspartate (NMDA)-induced excitotoxicity, a noxious process which decreases cellular viability, alters gene expression and promotes excessive mROS production. Incubation of the neuronal cell line SH-SY5Y with NMDA decreased cellular viability and increased mitochondrial superoxide production; pre-incubation with ASX prevented these effects. Additionally, incubation of SH-SY5Y cells with ASX effectively reduced the basal mROS production and prevented hydrogen peroxide-induced cell death. In primary hippocampal neurons, transfected with a genetically encoded cytoplasmic Ca2+ sensor, ASX also prevented the increase in intracellular Ca2+ concentration induced by NMDA. We suggest that, by preventing the noxious mROS and Ca2+ increases that occur under excitotoxic conditions, ASX could be useful as a therapeutic agent in neurodegenerative pathologies that involve alterations in Ca2+ homeostasis and ROS generation.
Collapse
Affiliation(s)
- Francisca García
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Pedro Lobos
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (P.L.); (A.P.-L.); (C.H.)
| | - Alejandra Ponce
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Karla Cataldo
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Daniela Meza
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Patricio Farías
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Carolina Estay
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Felipe Oyarzun-Ampuero
- Department of Technology and Pharmaceutical Sciences, Faculty of Chemical and Pharmaceutical Sciences, Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile;
| | - Rodrigo Herrera-Molina
- Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany;
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370854, Chile
| | - Andrea Paula-Lima
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (P.L.); (A.P.-L.); (C.H.)
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago 8380000, Chile
| | - Álvaro O. Ardiles
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Interdisciplinary Center of Neuroscience of Valparaíso, Universidad de Valparaíso, Valparaíso 2381850, Chile
- Interdisciplinary Center for Health Studies, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Cecilia Hidalgo
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile; (P.L.); (A.P.-L.); (C.H.)
- Department of Neurosciences and Program of Physiology and Biophysics, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile
- Center for Exercise, Metabolism and Cancer Studies, Faculty of Medicine, Universidad de Chile, Santiago 8380000, Chile
| | - Tatiana Adasme
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370854, Chile
- Correspondence: (T.A.); (P.M.); Tel.: +56-29-786-496 (T.A.); +56-32-250-7368 (P.M.)
| | - Pablo Muñoz
- Laboratory of Cellular and Molecular Plasticity, Department of Pathology and Physiology, Medical School, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile; (F.G.); (A.P.); (K.C.); (D.M.); (P.F.); (C.E.); (Á.O.A.)
- Translational Neurology Center, Faculty of Medicine, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Biomedical Research Center, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Correspondence: (T.A.); (P.M.); Tel.: +56-29-786-496 (T.A.); +56-32-250-7368 (P.M.)
| |
Collapse
|
31
|
Liu Y, Xu S, Bian H, Qian Y, Li H, Shu S, Chen J, Cao X, Gu Y, Jin J, Zhang X, Xu Y, Zhu X. Xingnaojing ameliorates synaptic plasticity and memory deficits in an Aβ 1-42 induced mouse model of Alzheimer's disease. J Pharmacol Sci 2020; 143:245-254. [PMID: 32482409 DOI: 10.1016/j.jphs.2020.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/11/2020] [Accepted: 03/23/2020] [Indexed: 11/26/2022] Open
Abstract
The accumulation of insoluble amyloid β (Aβ) peptides is one of the pathological changes in Alzheimer's disease (AD), which induced synaptic plasticity impairment and excitatory amino acid toxicity associated with decreased memory function. Xingnaojing (XNJ), a well-known prescription in traditional Chinese medicine, has been used for the treatment of stroke for many years in China. In this study, we aim to investigate the therapeutic effects of XNJ in a hippocampus of Aβ1-42 induced mouse model of AD which showed significant memory loss and impaired synaptic morphology and function. Treatment of XNJ could attenuate spatial and working memory dysfunction, increase dendritic spine density and improve long-term potential (LTP) induction. In addition, XNJ treatment significantly increased the level of N-methyl-d-aspartate receptors (NMDARs) and inhibit the NMDA/α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) ratio in AD mice. XNJ treatment also activated the AKT/mechanistic target of rapamycin (mTOR) pathway, while inhibition of the mTOR pathway by rapamycin could reverse the protective effects of XNJ treatment. In conclusion, XNJ protected against synaptic plasticity and memory impairment in AD mice via the activation of AKT/mTOR signaling pathway, suggesting XNJ as an alternative treatment for AD.
Collapse
Affiliation(s)
- Yi Liu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Siyi Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Huijie Bian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yi Qian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Huiya Li
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Shu Shu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Jiang Chen
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xiang Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yue Gu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Jiali Jin
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Xi Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
| | - Xiaolei Zhu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
32
|
Marcantoni A, Cerullo MS, Buxeda P, Tomagra G, Giustetto M, Chiantia G, Carabelli V, Carbone E. Amyloid Beta42 oligomers up-regulate the excitatory synapses by potentiating presynaptic release while impairing postsynaptic NMDA receptors. J Physiol 2020; 598:2183-2197. [PMID: 32246769 DOI: 10.1113/jp279345] [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] [Received: 11/25/2019] [Accepted: 03/26/2020] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS NMDA receptors (NMDARs) are key molecules for controlling neuronal plasticity, learning and memory processes. Their function is impaired during Alzheimer's disease (AD) but the exact consequence on synaptic function is not yet fully identified. An important hallmark of AD onset is represented by the neuronal accumulation of Amyloid Beta42 oligomers (Abeta42) that we have recently shown to be responsible for the increased intracellular Ca2+ concentration through ryanodine receptors (RyRs). Here we characterized the effects of Abeta42 on NMDA synapses showing specific pre- and post-synaptic functional changes that lead to a potentiation of basal and synchronous NMDA synaptic transmission. These overall effects can be abolished by decreasing Ca2+ release from RyRs with specific inhibitors that we propose as new pharmacological tools for AD treatment. ABSTRACT We have recently shown that Amyloid Beta42 oligomers (Abeta42) cause calcium dysregulation in hippocampal neurons by stimulating Ca2+ release from ryanodine receptors (RyRs) and inhibiting Ca2+ entry through NMDA receptors (NMDARs). Here, we found that Abeta42 decrease the average NMDA-activated inward current and that Ca2+ entry through NMDARs is accompanied by Ca2+ release from the stores. The overall amount of intraellular Ca2+ concentration([Ca2+ ]i ) increase during NMDA application is 50% associated with RyR opening and 50% with NMDARs activation. Addition of Abeta42 does not change this proportion. We estimated the number of NMDARs expressed in hippocampal neurons and their unitary current. We found that Abeta42 decrease the number of NMDARs without altering their unitary current. Paradoxically, the oligomer increases the size of electrically evoked eEPSCs induced by NMDARs activation. We found that this is the consequence of the increased release probability (p) of glutamate and the number of release sites (N) of NMDA synapses, while the quantal size (q) is significantly decreased as expected from the decreased number of NMDARs. An increased number of release sites induced by Abeta42 is also supported by the increased size of the ready releasable pool (RRPsyn) and by the enhanced percentage of paired pulse depression (PPD). Interestingly, the RyRs inhibitor dantrolene prevents the increase of PPD induced by Abeta42 oligomers. In conclusion, Abeta42 up-regulates NMDA synaptic responses with a mechanism involving RyRs that occurs during the early stages of Alzheimer's disease (AD) onset. This suggests that new selective modulators of RyRs may be useful for designing effective therapies to treat AD patients.
Collapse
Affiliation(s)
| | | | - Pol Buxeda
- Department of Drug Science and Technology, Torino University, Italy
| | - Giulia Tomagra
- Department of Drug Science and Technology, Torino University, Italy
| | - Maurizio Giustetto
- Department of Neurosciences / National Institute of Neuroscience, Torino University, Italy.,National Institute of Neuroscience-Italy, Turin, Italy
| | | | | | - Emilio Carbone
- Department of Drug Science and Technology, Torino University, Italy
| |
Collapse
|
33
|
Findley CA, Bartke A, Hascup KN, Hascup ER. Amyloid Beta-Related Alterations to Glutamate Signaling Dynamics During Alzheimer's Disease Progression. ASN Neuro 2020; 11:1759091419855541. [PMID: 31213067 PMCID: PMC6582288 DOI: 10.1177/1759091419855541] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) ranks sixth on the Centers for Disease Control and Prevention Top 10 Leading Causes of Death list for 2016, and the Alzheimer’s Association attributes 60% to 80% of dementia cases as AD related. AD pathology hallmarks include accumulation of senile plaques and neurofibrillary tangles; however, evidence supports that soluble amyloid beta (Aβ), rather than insoluble plaques, may instigate synaptic failure. Soluble Aβ accumulation results in depression of long-term potentiation leading to cognitive deficits commonly characterized in AD. The mechanisms through which Aβ incites cognitive decline have been extensively explored, with a growing body of evidence pointing to modulation of the glutamatergic system. The period of glutamatergic hypoactivation observed alongside long-term potentiation depression and cognitive deficits in later disease stages may be the consequence of a preceding period of increased glutamatergic activity. This review will explore the Aβ-related changes to the tripartite glutamate synapse resulting in altered cell signaling throughout disease progression, ultimately culminating in oxidative stress, synaptic dysfunction, and neuronal loss.
Collapse
Affiliation(s)
- Caleigh A Findley
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- 3 Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.,4 Department of Molecular Biology, Microbiology & Biochemistry, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
34
|
Wang Y, Liang G, Liang S, Mund R, Shi Y, Wei H. Dantrolene Ameliorates Impaired Neurogenesis and Synaptogenesis in Induced Pluripotent Stem Cell Lines Derived from Patients with Alzheimer's Disease. Anesthesiology 2020; 132:1062-1079. [PMID: 32149777 PMCID: PMC7160009 DOI: 10.1097/aln.0000000000003224] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Overactivation of ryanodine receptors and the resulting impaired calcium homeostasis contribute to Alzheimer's disease-related pathophysiology. This study hypothesized that exposing neuronal progenitors derived from induced pluripotent stems cells of patients with Alzheimer's disease to dantrolene will increase survival, proliferation, neurogenesis, and synaptogenesis. METHODS Induced pluripotent stem cells obtained from skin fibroblast of healthy subjects and patients with familial and sporadic Alzheimer's disease were used. Biochemical and immunohistochemical methods were applied to determine the effects of dantrolene on the viability, proliferation, differentiation, and calcium dynamics of these cells. RESULTS Dantrolene promoted cell viability and proliferation in these two cell lines. Compared with the control, differentiation into basal forebrain cholinergic neurons significantly decreased by 10.7% (32.9 ± 3.6% vs. 22.2 ± 2.6%, N = 5, P = 0.004) and 9.2% (32.9 ± 3.6% vs. 23.7 ± 3.1%, N = 5, P = 0.017) in cell lines from sporadic and familial Alzheimer's patients, respectively, which were abolished by dantrolene. Synapse density was significantly decreased in cortical neurons generated from stem cells of sporadic Alzheimer's disease by 58.2% (237.0 ± 28.4 vs. 99.0 ± 16.6 arbitrary units, N = 4, P = 0.001) or familial Alzheimer's disease by 52.3% (237.0 ± 28.4 vs.113.0 ± 34.9 vs. arbitrary units, N = 5, P = 0.001), which was inhibited by dantrolene in the familial cell line. Compared with the control, adenosine triphosphate (30 µM) significantly increased higher peak elevation of cytosolic calcium concentrations in the cell line from sporadic Alzheimer's patients (84.1 ± 27.0% vs. 140.4 ± 40.2%, N = 5, P = 0.049), which was abolished by the pretreatment of dantrolene. Dantrolene inhibited the decrease of lysosomal vacuolar-type H-ATPase and the impairment of autophagy activity in these two cell lines from Alzheimer's disease patients. CONCLUSIONS Dantrolene ameliorated the impairment of neurogenesis and synaptogenesis, in association with restoring intracellular Ca homeostasis and physiologic autophagy, cell survival, and proliferation in induced pluripotent stem cells and their derived neurons from sporadic and familial Alzheimer's disease patients.
Collapse
Affiliation(s)
- Yong Wang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Anesthesiology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuqing Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Anesthesiology, the First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Rachel Mund
- Undergraduate Student, College of Art and Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yun Shi
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Anesthesiology, Children’s hospital of Fudan University, Shanghai, 201102, China
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
Intracellular Calcium Responses Encode Action Potential Firing in Spinal Cord Lamina I Neurons. J Neurosci 2020; 40:4439-4456. [PMID: 32341097 DOI: 10.1523/jneurosci.0206-20.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/05/2020] [Accepted: 04/19/2020] [Indexed: 12/19/2022] Open
Abstract
Maladaptive plasticity of neurons in lamina I of the spinal cord is a lynchpin for the development of chronic pain, and is critically dependent on intracellular calcium signaling. However, the relationship between neuronal activity and intracellular calcium in these neurons is unknown. Here we combined two-photon calcium imaging with whole-cell electrophysiology to determine how action potential firing drives calcium responses within subcellular compartments of male rat spinal cord lamina I neurons. We found that single action potentials generated at the soma increase calcium concentration in the somatic cytosol and nucleus, and these calcium responses invade dendrites and dendritic spines by active backpropagation. Calcium responses in each compartment were dependent on voltage-gated calcium channels, and somatic and nuclear calcium responses were amplified by release of calcium from ryanodine-sensitive intracellular stores. Grouping single action potential-evoked calcium responses by neuron type demonstrated their presence in all defined types, as well as a high degree of similarity in calcium responses between neuron types. With bursts of action potentials, we found that calcium responses have the capacity to encode action potential frequency and number in all compartments, with action potential number being preferentially encoded. Together, these findings indicate that intracellular calcium serves as a readout of neuronal activity within lamina I neurons, providing a unifying mechanism through which activity may regulate plasticity, including that seen in chronic pain.SIGNIFICANCE STATEMENT Despite their critical role in both acute pain sensation and chronic pain, little is known of the fundamental physiology of spinal cord lamina I neurons. This is especially the case with respect to calcium dynamics within these neurons, which could regulate maladaptive plasticity observed in chronic pain. By combining two-photon calcium imaging and patch-clamp electrophysiological recordings from lamina I neurons, we found that action potential firing induces calcium responses within the somatic cytosol, nucleus, dendrites, and dendritic spines of lamina I neurons. Our findings demonstrate the presence of actively backpropagating action potentials, shifting our understanding of how these neurons process information, such that calcium provides a mechanism for lamina I neurons to track their own activity.
Collapse
|
36
|
Kawano H, Mitchell SB, Koh JY, Goodman KM, Harata NC. Calcium-induced calcium release in noradrenergic neurons of the locus coeruleus. Brain Res 2020; 1729:146627. [PMID: 31883849 DOI: 10.1016/j.brainres.2019.146627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 12/11/2022]
Abstract
The locus coeruleus (LC) is a nucleus within the brainstem that consists of norepinephrine-releasing neurons. It is involved in broad processes including cognitive and emotional functions. Understanding the mechanisms that control the excitability of LC neurons is important because they innervate widespread brain regions. One of the key regulators is cytosolic calcium concentration ([Ca2+]c), the increases in which can be amplified by calcium-induced calcium release (CICR) from intracellular calcium stores. Although the electrical activities of LC neurons are regulated by changes in [Ca2+]c, the extent of CICR involvement in this regulation has remained unclear. Here we show that CICR hyperpolarizes acutely dissociated LC neurons of the rat and demonstrate the underlying pathway. When CICR was activated by extracellular application of 10 mM caffeine, LC neurons were hyperpolarized in the current-clamp mode of patch-clamp recording, and the majority of neurons showed an outward current in the voltage-clamp mode. This outward current was accompanied by increased membrane conductance, and its reversal potential was close to the K+ equilibrium potential, indicating that it is mediated by opening of K+ channels. The outward current was generated in the absence of extracellular calcium and was blocked when the calcium stores were inhibited by applying ryanodine. Pharmacological blockers indicated that it was mediated by Ca2+-activated K+ channels of the non-small conductance type. The application of caffeine increased [Ca2+]c, as visualized by fluorescence microscopy. These findings show CICR suppresses LC neuronal activity, and indicate its dynamic role in modulating the LC-mediated noradrenergic tone in the brain.
Collapse
Affiliation(s)
- Hiroyuki Kawano
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sara B Mitchell
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jin-Young Koh
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Molecular Otolaryngology and Renal Research Laboratories, Department of Otolaryngology-Head and Neck Surgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Biomedical Engineering, University of Iowa College of Engineering, Iowa City, IA, USA
| | - Kirsty M Goodman
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Biology & Biochemistry, University of Bath, Bath, UK
| | - N Charles Harata
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
37
|
Schrank S, McDaid J, Briggs CA, Mustaly-Kalimi S, Brinks D, Houcek A, Singer O, Bottero V, Marr RA, Stutzmann GE. Human-Induced Neurons from Presenilin 1 Mutant Patients Model Aspects of Alzheimer's Disease Pathology. Int J Mol Sci 2020; 21:ijms21031030. [PMID: 32033164 PMCID: PMC7037274 DOI: 10.3390/ijms21031030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/01/2020] [Accepted: 02/02/2020] [Indexed: 12/20/2022] Open
Abstract
Traditional approaches to studying Alzheimer’s disease (AD) using mouse models and cell lines have advanced our understanding of AD pathogenesis. However, with the growing divide between model systems and clinical therapeutic outcomes, the limitations of these approaches are increasingly apparent. Thus, to generate more clinically relevant systems that capture pathological cascades within human neurons, we generated human-induced neurons (HiNs) from AD and non-AD individuals to model cell autonomous disease properties. We selected an AD patient population expressing mutations in presenilin 1 (mPS1), which is linked to increased amyloid production, tau pathology, and calcium signaling abnormalities, among other features. While these AD components are detailed in model systems, they have yet to be collectively identified in human neurons. Thus, we conducted molecular, immune-based, electrophysiological, and calcium imaging studies to establish patterns of cellular pathology in this patient population. We found that mPS1 HiNs generate increased Aβ42 and hyperphosphorylated tau species relative to non-AD controls, and exaggerated ER calcium responses that are normalized with ryanodine receptor (RyR) negative allosteric modulators. The inflammasome product, interleukin-18 (IL-18), also increased PS1 expression. This work highlights the potential for HiNs to model AD pathology and validates their role in defining cellular pathogenesis and their utility for therapeutic screening.
Collapse
Affiliation(s)
- Sean Schrank
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; (S.S.); (J.M.); (C.A.B.); (S.M.-K.); (V.B.)
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - John McDaid
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; (S.S.); (J.M.); (C.A.B.); (S.M.-K.); (V.B.)
| | - Clark A. Briggs
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; (S.S.); (J.M.); (C.A.B.); (S.M.-K.); (V.B.)
| | - Sarah Mustaly-Kalimi
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; (S.S.); (J.M.); (C.A.B.); (S.M.-K.); (V.B.)
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Deanna Brinks
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd. North, Chicago, IL 60064, USA;
| | - Aiden Houcek
- Lake Forest College, Lake Forest, IL 60045, USA;
| | - Oded Singer
- Weizmann Institute of Science, Life Sciences Core Facilities, Rehovot 76100, Israel;
| | - Virginie Bottero
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; (S.S.); (J.M.); (C.A.B.); (S.M.-K.); (V.B.)
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd. North, Chicago, IL 60064, USA;
| | - Robert A. Marr
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; (S.S.); (J.M.); (C.A.B.); (S.M.-K.); (V.B.)
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd. North, Chicago, IL 60064, USA;
- Correspondence: (R.A.M.); (G.E.S.)
| | - Grace E. Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA; (S.S.); (J.M.); (C.A.B.); (S.M.-K.); (V.B.)
- School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd. North, Chicago, IL 60064, USA;
- Correspondence: (R.A.M.); (G.E.S.)
| |
Collapse
|
38
|
Popugaeva E, Bezprozvanny I, Chernyuk D. Reversal of Calcium Dysregulation as Potential Approach for Treating Alzheimer's Disease. Curr Alzheimer Res 2020; 17:344-354. [PMID: 32469698 PMCID: PMC8210816 DOI: 10.2174/1567205017666200528162046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 02/25/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Despite decades of research and effort, there is still no effective disease-modifying treatment for Alzheimer's Disease (AD). Most of the recent AD clinical trials were targeting amyloid pathway, but all these trials failed. Although amyloid pathology is a hallmark and defining feature of AD, targeting the amyloid pathway has been very challenging due to low efficacy and serious side effects. Alternative approaches or mechanisms for our understanding of the major cause of memory loss in AD need to be considered as potential therapeutic targets. Increasing studies suggest that Ca2+ dysregulation in AD plays an important role in AD pathology and is associated with other AD abnormalities, such as excessive inflammation, increased ROS, impaired autophagy, neurodegeneration, synapse, and cognitive dysfunction. Ca2+ dysregulation in cytosolic space, Endoplasmic Reticulum (ER) and mitochondria have been reported in the context of various AD models. Drugs or strategies, to correct the Ca2+ dysregulation in AD, have been demonstrated to be promising as an approach for the treatment of AD in preclinical models. This review will discuss the mechanisms of Ca2+ dysregulation in AD and associated pathology and discuss potential approaches or strategies to develop novel drugs for the treatment of AD by targeting Ca2+ dysregulation.
Collapse
Affiliation(s)
- Elena Popugaeva
- Department of Medical Physics, Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg Polytechnic University, St Petersburg, Russia
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center, Dallas, USA
| | - Daria Chernyuk
- Department of Medical Physics, Laboratory of Molecular Neurodegeneration, Peter the Great St Petersburg Polytechnic University, St Petersburg, Russia
| |
Collapse
|
39
|
Presenilin 1 Regulates [Ca 2+]i and Mitochondria/ER Interaction in Cultured Rat Hippocampal Neurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7284967. [PMID: 31467635 PMCID: PMC6701405 DOI: 10.1155/2019/7284967] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/26/2019] [Accepted: 06/11/2019] [Indexed: 01/17/2023]
Abstract
Mutations in the presenilin 1 (PS1) gene are a major trigger of familial Alzheimer's disease (AD), yet the mechanisms affected by mutated PS1 causing cognitive decline are not yet elucidated. In the present study, we compared rat hippocampal neurons in culture, transfected with PS1 or with mutant (M146V) PS1 (mPS1) plasmids in several neuronal functions. Initially, we confirmed earlier observations that mPS1-expressing neurons are endowed with fewer mature “mushroom” spines and more filopodial immature protrusions. The correlation between calcium changes in the cytosol, mitochondria, and endoplasmic reticulum (ER) is mitigated in the mPS1 neurons, tested by the response to an abrupt increase in ambient [Ca2+]o; cytosolic [Ca2+]i is higher in the mPS1 neurons but mitochondrial [Ca2+] is lower than in control neurons. Strikingly, mPS1-transfected neurons express higher excitability and eventual lower survival rate when exposed to the oxidative stressor, paraquat. These results highlight an impaired calcium regulation in mPS1 neurons, resulting in a reduced ability to handle oxidative stress, which may lead to cell death and AD.
Collapse
|
40
|
Lin CH, Lane HY. The Role of N-Methyl-D-Aspartate Receptor Neurotransmission and Precision Medicine in Behavioral and Psychological Symptoms of Dementia. Front Pharmacol 2019; 10:540. [PMID: 31191302 PMCID: PMC6539199 DOI: 10.3389/fphar.2019.00540] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 04/29/2019] [Indexed: 12/16/2022] Open
Abstract
While the world’s population is aging, the prevalence of dementia and the associated behavioral and psychological symptoms of dementia (BPSD) rises rapidly. BPSD are associated with worsening of cognitive function and poorer prognosis. No pharmacological treatment has been approved to be beneficial for BPSD to date. Dysfunction of the N-methyl-D-aspartate receptor (NMDAR)-related neurotransmission leads to cognitive impairment and behavioral changes, both of which are core symptoms of BPSD. Memantine, an NMDAR partial antagonist, is used to treat moderate to severe Alzheimer’s disease (AD). On the other hand, a D-amino acid oxidase inhibitor improved early-phase AD. Whether to enhance or to attenuate the NMDAR may depend on the phases of dementia. It will be valuable to develop biomarkers indicating the activity of NMDAR, particularly in BPSD. In addition, recent reports suggest that gender difference exists in the treatment of dementia. Selecting subpopulations of patients with BPSD who are prone to improvement with treatment would be important. We reviewed literatures regarding the treatment of BPSD, focusing on the NMDAR-related modulation and precision medicine. Future studies examining the NMDAR modulators with the aid of potential biomarkers to tailor the treatment for individualized patients with BPSD are warranted.
Collapse
Affiliation(s)
- Chieh-Hsin Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Psychiatry and Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan.,Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
| |
Collapse
|
41
|
Gavello D, Calorio C, Franchino C, Cesano F, Carabelli V, Carbone E, Marcantoni A. Early Alterations of Hippocampal Neuronal Firing Induced by Abeta42. Cereb Cortex 2019; 28:433-446. [PMID: 27999123 DOI: 10.1093/cercor/bhw377] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Indexed: 12/11/2022] Open
Abstract
We studied the effect of Amyloid β 1-42 oligomers (Abeta42) on Ca2+ dependent excitability profile of hippocampal neurons. Abeta42 is one of the Amyloid beta peptides produced by the proteolytic processing of the amyloid precursor protein and participates in the initiating event triggering the progressive dismantling of synapses and neuronal circuits. Our experiments on cultured hippocampal network reveal that Abeta42 increases intracellular Ca2+ concentration by 46% and inhibits firing discharge by 19%. More precisely, Abeta42 differently regulates ryanodine (RyRs), NMDA receptors (NMDARs), and voltage gated calcium channels (VGCCs) by increasing Ca2+ release through RyRs and inhibiting Ca2+ influx through NMDARs and VGCCs. The overall increased intracellular Ca2+ concentration causes stimulation of K+ current carried by big conductance Ca2+ activated potassium (BK) channels and hippocampal network firing inhibition. We conclude that Abeta42 alters neuronal function by means of at least 4 main targets: RyRs, NMDARs, VGCCs, and BK channels. The development of selective modulators of these channels may in turn be useful for developing effective therapies that could enhance the quality of life of AD patients during the early onset of the pathology.
Collapse
Affiliation(s)
- Daniela Gavello
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Chiara Calorio
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Claudio Franchino
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Federico Cesano
- Department of Chemistry Via Pietro Giuria 7, Torino University, 10125 Torino, Italy
| | - Valentina Carabelli
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Emilio Carbone
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| | - Andrea Marcantoni
- Department of Drug Science and Technology, Torino University, Corso Raffaello 30, 10125 Torino, Italy
| |
Collapse
|
42
|
Chakroborty S, Hill ES, Christian DT, Helfrich R, Riley S, Schneider C, Kapecki N, Mustaly-Kalimi S, Seiler FA, Peterson DA, West AR, Vertel BM, Frost WN, Stutzmann GE. Reduced presynaptic vesicle stores mediate cellular and network plasticity defects in an early-stage mouse model of Alzheimer's disease. Mol Neurodegener 2019; 14:7. [PMID: 30670054 PMCID: PMC6343260 DOI: 10.1186/s13024-019-0307-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/13/2019] [Indexed: 01/27/2023] Open
Abstract
Background Identifying effective strategies to prevent memory loss in AD has eluded researchers to date, and likely reflects insufficient understanding of early pathogenic mechanisms directly affecting memory encoding. As synaptic loss best correlates with memory loss in AD, refocusing efforts to identify factors driving synaptic impairments may provide the critical insight needed to advance the field. In this study, we reveal a previously undescribed cascade of events underlying pre and postsynaptic hippocampal signaling deficits linked to cognitive decline in AD. These profound alterations in synaptic plasticity, intracellular Ca2+ signaling, and network propagation are observed in 3–4 month old 3xTg-AD mice, an age which does not yet show overt histopathology or major behavioral deficits. Methods In this study, we examined hippocampal synaptic structure and function from the ultrastructural level to the network level using a range of techniques including electron microscopy (EM), patch clamp and field potential electrophysiology, synaptic immunolabeling, spine morphology analyses, 2-photon Ca2+ imaging, and voltage-sensitive dye-based imaging of hippocampal network function in 3–4 month old 3xTg-AD and age/background strain control mice. Results In 3xTg-AD mice, short-term plasticity at the CA1-CA3 Schaffer collateral synapse is profoundly impaired; this has broader implications for setting long-term plasticity thresholds. Alterations in spontaneous vesicle release and paired-pulse facilitation implicated presynaptic signaling abnormalities, and EM analysis revealed a reduction in the ready-releasable and reserve pools of presynaptic vesicles in CA3 terminals; this is an entirely new finding in the field. Concurrently, increased synaptically-evoked Ca2+ in CA1 spines triggered by LTP-inducing tetani is further enhanced during PTP and E-LTP epochs, and is accompanied by impaired synaptic structure and spine morphology. Notably, vesicle stores, synaptic structure and short-term plasticity are restored by normalizing intracellular Ca2+ signaling in the AD mice. Conclusions These findings suggest the Ca2+ dyshomeostasis within synaptic compartments has an early and fundamental role in driving synaptic pathophysiology in early stages of AD, and may thus reflect a foundational disease feature driving later cognitive impairment. The overall significance is the identification of previously unidentified defects in pre and postsynaptic compartments affecting synaptic vesicle stores, synaptic plasticity, and network propagation, which directly impact memory encoding. Electronic supplementary material The online version of this article (10.1186/s13024-019-0307-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shreaya Chakroborty
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Evan S Hill
- Department of Cell Biology and Anatomy, The Chicago Medical School; Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Daniel T Christian
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Rosalind Helfrich
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Shannon Riley
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Corinne Schneider
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Nicolas Kapecki
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Sarah Mustaly-Kalimi
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Figen A Seiler
- Electron Microscopy Center, RFUMS, North Chicago, IL, 60064, USA
| | - Daniel A Peterson
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Anthony R West
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Barbara M Vertel
- Department of Cell Biology and Anatomy, The Chicago Medical School; Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA.,Electron Microscopy Center, RFUMS, North Chicago, IL, 60064, USA
| | - William N Frost
- Department of Cell Biology and Anatomy, The Chicago Medical School; Center for Brain Function and Repair, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA
| | - Grace E Stutzmann
- Department of Neuroscience, The Chicago Medical School; The Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL, 60064, USA.
| |
Collapse
|
43
|
Shi Y, Wang Y, Wei H. Dantrolene : From Malignant Hyperthermia to Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2019; 18:668-676. [PMID: 29921212 PMCID: PMC7754833 DOI: 10.2174/1871527317666180619162649] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 05/07/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022]
Abstract
Dantrolene, a ryanodine receptor antagonist, is primarily known as the only clinically acceptable and effective treatment for Malignant Hyperthermia (MH). Inhibition of Ryanodine Receptor (RyR) by dantrolene decreases the abnormal calcium release from the Sarcoplasmic Reticulum (SR) or Endoplasmic Reticulum (ER), where RyR is located. Recently, emerging researches on dissociated cells, brains slices, live animal models and patients have demonstrated that altered RyR expression and function can also play a vital role in the pathogenesis of Alzheimer's Disease (AD). Therefore, dantrolene is now widely studied as a novel treatment for AD, targeting the blockade of RyR channels or another alternative pathway, such as the inhibitory effects of NMDA glutamate receptors and the effects of ER-mitochondria connection. However, the therapeutic effects are not consistent. In this review, we focus on the relationship between the altered RyR expression and function and the pathogenesis of AD, and the potential application of dantrolene as a novel treatment for the disease.
Collapse
Affiliation(s)
- Yun Shi
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104, USA
- Department of Anesthesiology, Children’s Hospital of Fudan University, Shanghai, China
| | - Yong Wang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104, USA
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104, USA
| |
Collapse
|
44
|
Asavapanumas N, Brawek B, Martus P, Garaschuk O. Role of intracellular Ca2+ stores for an impairment of visual processing in a mouse model of Alzheimer's disease. Neurobiol Dis 2019; 121:315-326. [DOI: 10.1016/j.nbd.2018.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/28/2018] [Accepted: 10/22/2018] [Indexed: 12/21/2022] Open
|
45
|
More J, Galusso N, Veloso P, Montecinos L, Finkelstein JP, Sanchez G, Bull R, Valdés JL, Hidalgo C, Paula-Lima A. N-Acetylcysteine Prevents the Spatial Memory Deficits and the Redox-Dependent RyR2 Decrease Displayed by an Alzheimer's Disease Rat Model. Front Aging Neurosci 2018; 10:399. [PMID: 30574085 PMCID: PMC6291746 DOI: 10.3389/fnagi.2018.00399] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/19/2018] [Indexed: 12/28/2022] Open
Abstract
We have previously reported that primary hippocampal neurons exposed to synaptotoxic amyloid beta oligomers (AβOs), which are likely causative agents of Alzheimer’s disease (AD), exhibit abnormal Ca2+ signals, mitochondrial dysfunction and defective structural plasticity. Additionally, AβOs-exposed neurons exhibit a decrease in the protein content of type-2 ryanodine receptor (RyR2) Ca2+ channels, which exert critical roles in hippocampal synaptic plasticity and spatial memory processes. The antioxidant N-acetylcysteine (NAC) prevents these deleterious effects of AβOs in vitro. The main contribution of the present work is to show that AβOs injections directly into the hippocampus, by engaging oxidation-mediated reversible pathways significantly decreased RyR2 protein content but increased single RyR2 channel activation by Ca2+ and caused considerable spatial memory deficits. AβOs injections into the CA3 hippocampal region impaired rat performance in the Oasis maze spatial memory task, decreased hippocampal glutathione levels and overall content of plasticity-related proteins (c-Fos, Arc, and RyR2) and increased ERK1/2 phosphorylation. In contrast, in hippocampus-derived mitochondria-associated membranes (MAM) AβOs injections increased RyR2 levels. Rats fed with NAC for 3-weeks prior to AβOs injections displayed comparable redox potential, RyR2 and Arc protein contents, similar ERK1/2 phosphorylation and RyR2 single channel activation by Ca2+ as saline-injected (control) rats. NAC-fed rats subsequently injected with AβOs displayed the same behavior in the spatial memory task as control rats. Based on the present in vivo results, we propose that redox-sensitive neuronal RyR2 channels partake in the mechanism underlying AβOs-induced memory disruption in rodents.
Collapse
Affiliation(s)
- Jamileth More
- Faculty of Medicine, Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile
| | - Nadia Galusso
- Department of Neurochemistry, Stockholm University, Stockholm, Sweden
| | - Pablo Veloso
- Faculty of Dentistry, Institute for Research in Dental Sciences, Universidad de Chile, Santiago, Chile
| | - Luis Montecinos
- CEMC, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Gina Sanchez
- CEMC, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Pathophysiology Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Ricardo Bull
- Physiology and Biophysics Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - José Luis Valdés
- Faculty of Medicine, Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cecilia Hidalgo
- Faculty of Medicine, Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile.,CEMC, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Physiology and Biophysics Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrea Paula-Lima
- Faculty of Medicine, Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile.,Faculty of Dentistry, Institute for Research in Dental Sciences, Universidad de Chile, Santiago, Chile
| |
Collapse
|
46
|
Mustaly-Kalimi S, Littlefield AM, Stutzmann GE. Calcium Signaling Deficits in Glia and Autophagic Pathways Contributing to Neurodegenerative Disease. Antioxid Redox Signal 2018; 29:1158-1175. [PMID: 29634342 DOI: 10.1089/ars.2017.7266] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Numerous cellular processes and signaling mechanisms have been identified that contribute to Alzheimer's disease (AD) pathology; however, a comprehensive or unifying pathway that binds together the major disease features remains elusive. As an upstream mechanism, altered calcium (Ca2+) signaling is a common driving force for many pathophysiological events that emerge during normal aging and development of neurodegenerative disease. Recent Advances: Over the previous three decades, accumulated evidence has validated the concept that intracellular Ca2+ dysregulation is centrally involved in AD pathogenesis, including the aggregation of pathogenic β-amyloid (Aβ) and phospho-τ species, synapse loss and dysfunction, cognitive impairment, and neurotoxicity. CRITICAL ISSUES Although neuronal Ca2+ signaling within the cytosol and endoplasmic reticulum (ER) has been well studied, other critical central nervous system-resident cell types affected by aberrant Ca2+ signaling, such as astrocytes and microglia, have not been considered as thoroughly. In addition, certain intracellular Ca2+-harboring organelles have been well studied, such as the ER and mitochondria; however other critical Ca2+-regulated organelles, such as lysosomes and autophagosomes, have only more recently been investigated. In this review, we examine Ca2+ dysregulation in microglia and astrocytes, as well as key intracellular organelles important for cellular maintenance and protein handling. Ca2+ dysregulation within these non-neuronal cells and organelles is hypothesized to disrupt the effective clearance of misaggregated proteins and cellular signaling pathways needed for memory networks. FUTURE DIRECTIONS Overall, we aim to explore how these disrupted mechanisms could be involved in AD pathology and consider their role as potential therapeutic targets. Antioxid. Redox Signal. 29, 1158-1175.
Collapse
Affiliation(s)
- Sarah Mustaly-Kalimi
- 1 Department of Neuroscience, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science , North Chicago, Illinois
| | - Alyssa M Littlefield
- 1 Department of Neuroscience, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science , North Chicago, Illinois
| | - Grace E Stutzmann
- 2 Department of Neuroscience, The Chicago Medical School, Rosalind Franklin University of Medicine and Science , North Chicago, Illinois
| |
Collapse
|
47
|
Popugaeva E, Pchitskaya E, Bezprozvanny I. Dysregulation of Intracellular Calcium Signaling in Alzheimer's Disease. Antioxid Redox Signal 2018; 29:1176-1188. [PMID: 29890840 PMCID: PMC6157344 DOI: 10.1089/ars.2018.7506] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
SIGNIFICANCE Calcium (Ca2+) hypothesis of Alzheimer's disease (AD) gains popularity. It points to new signaling pathways that may underlie AD pathogenesis. Based on calcium hypothesis, novel targets for the development of potential AD therapies are identified. Recent Advances: Recently, the key role of neuronal store-operated calcium entry (nSOCE) in the development of AD has been described. Correct regulation of nSOCE is necessary for the stability of postsynaptic contacts to preserve the memory formation. Molecular identity of hippocampal nSOCE is defined. Perspective nSOCE-activating molecule, prototype of future anti-AD drugs, is described. CRITICAL ISSUES Endoplasmic reticulum Ca2+ overload happens in many but not in all AD models. The nSOCE targeting therapy described in this review may not be universally applicable. FUTURE DIRECTIONS There is a need to determine whether AD is a syndrome with one critical signaling pathway that initiates pathology, or it is a disorder with many different signaling pathways that are disrupted simultaneously or one after each other. It is necessary to validate applicability of nSOCE-activating therapy for the development of anti-AD medication. There is an experimental correlation between downregulated nSOCE and disrupted postsynaptic contacts in AD mouse models. Signaling mechanisms downstream of nSOCE which are responsible for the regulation of stability of postsynaptic contacts have to be discovered. That will bring new targets for the development of AD-preventing therapies. Antioxid. Redox Signal. 29, 1176-1188.
Collapse
Affiliation(s)
- Elena Popugaeva
- 1 Laboratory of Molecular Neurodegeneration, Department of Medical Physics, Peter the Great St.Petersburg Polytechnic University , St.Petersburg, Russian Federation
| | - Ekaterina Pchitskaya
- 1 Laboratory of Molecular Neurodegeneration, Department of Medical Physics, Peter the Great St.Petersburg Polytechnic University , St.Petersburg, Russian Federation
| | - Ilya Bezprozvanny
- 1 Laboratory of Molecular Neurodegeneration, Department of Medical Physics, Peter the Great St.Petersburg Polytechnic University , St.Petersburg, Russian Federation.,2 Department of Physiology, UT Southwestern Medical Center at Dallas , Dallas, Texas
| |
Collapse
|
48
|
Latulippe J, Lotito D, Murby D. A mathematical model for the effects of amyloid beta on intracellular calcium. PLoS One 2018; 13:e0202503. [PMID: 30133494 PMCID: PMC6105003 DOI: 10.1371/journal.pone.0202503] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 08/03/2018] [Indexed: 12/21/2022] Open
Abstract
The accumulation of Alzheimer's disease (AD) associated Amyloid beta (Aβ) oligomers can trigger aberrant intracellular calcium (Ca2+) levels by disrupting the intrinsic Ca2+ regulatory mechanism within cells. These disruptions can cause changes in homeostasis levels that can have detrimental effects on cell function and survival. Although studies have shown that Aβ can interfere with various Ca2+ fluxes, the complexity of these interactions remains elusive. We have constructed a mathematical model that simulates Ca2+ patterns under the influence of Aβ. Our simulations shows that Aβ can increase regions of mixed-mode oscillations leading to aberrant signals under various conditions. We investigate how Aβ affects individual flux contributions through inositol triphosphate (IP3) receptors, ryanodine receptors, and membrane pores. We demonstrate that controlling for the ryanodine receptor's maximal kinetic reaction rate may provide a biophysical way of managing aberrant Ca2+ signals. The influence of a dynamic model for IP3 production is also investigated under various conditions as well as the impact of changes in membrane potential. Our model is one of the first to investigate the effects of Aβ on a variety of cellular mechanisms providing a base modeling scheme from which further studies can draw on to better understand Ca2+ regulation in an AD environment.
Collapse
Affiliation(s)
- Joe Latulippe
- Mathematics Department, Norwich University, Northfield, Vermont, United States of America
- * E-mail:
| | - Derek Lotito
- Chemistry and Biochemistry Department, Norwich University, Northfield, Vermont, United States of America
| | - Donovan Murby
- Mathematics Department, Norwich University, Northfield, Vermont, United States of America
| |
Collapse
|
49
|
Russo R, Cattaneo F, Lippiello P, Cristiano C, Zurlo F, Castaldo M, Irace C, Borsello T, Santamaria R, Ammendola R, Calignano A, Miniaci MC. Motor coordination and synaptic plasticity deficits are associated with increased cerebellar activity of NADPH oxidase, CAMKII, and PKC at preplaque stage in the TgCRND8 mouse model of Alzheimer's disease. Neurobiol Aging 2018; 68:123-133. [DOI: 10.1016/j.neurobiolaging.2018.02.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/14/2018] [Accepted: 02/24/2018] [Indexed: 10/17/2022]
|
50
|
Tong BCK, Wu AJ, Li M, Cheung KH. Calcium signaling in Alzheimer's disease & therapies. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1745-1760. [PMID: 30059692 DOI: 10.1016/j.bbamcr.2018.07.018] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and is characterized by the accumulation of amyloid (Aβ) plaques and neurofibrillary tangles in the brain. Much attention has been given to develop AD treatments based on the amyloid cascade hypothesis; however, none of these drugs had good efficacy at improving cognitive functions in AD patients suggesting that Aβ might not be the disease origin. Thus, there are urgent needs for the development of new therapies that target on the proximal cause of AD. Cellular calcium (Ca2+) signals regulate important facets of neuronal physiology. An increasing body of evidence suggests that age-related dysregulation of neuronal Ca2+ homeostasis may play a proximal role in the pathogenesis of AD as disrupted Ca2+ could induce synaptic deficits and promote the accumulation of Aβ plaques and neurofibrillary tangles. Given that Ca2+ disruption is ubiquitously involved in all AD pathologies, it is likely that using chemical agents or small molecules specific to Ca2+ channels or handling proteins on the plasma membrane and membranes of intracellular organelles to correct neuronal Ca2+ dysregulation could open up a new approach to AD prevention and treatment. This review summarizes current knowledge on the molecular mechanisms linking Ca2+ dysregulation with AD pathologies and discusses the possibility of correcting neuronal Ca2+ disruption as a therapeutic approach for AD.
Collapse
Affiliation(s)
- Benjamin Chun-Kit Tong
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Aston Jiaxi Wu
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - King-Ho Cheung
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.
| |
Collapse
|