1
|
Du X, Wang H, Liu S, Song Y, Chen X, Chen Z, Zhou R, Du J, Zhang W, Gao R, Li H, Zhang G, Mao X, Chang L, Wu Y. Astrocytic GluN2A alleviates sleep deprivation-induced elevation of Aβ through regulating neprilysin and AQP4 via the calcineurin/NFAT pathway. Prog Neurobiol 2025; 248:102744. [PMID: 40032156 DOI: 10.1016/j.pneurobio.2025.102744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/22/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
Sleep disorders can increase amyloid beta (Aβ) burden in the brain and are linked to Alzheimer's disease (AD) risk. The precise mechanism by which sleep disturbances elevate Aβ levels is unclear. Our previous study has demonstrated that knocking down encoding gene Grin2a of astrocytic N-methyl-D-aspartate (NMDA) receptors GluN2A subunit could aggravate sleep deprivation (SD)-induced elevation of Aβ, indicating a protective role of astrocytic GluN2A in SD; but the underlying mechanism needs to be further elucidated. In our present study, using rat models of SD combined with specific astrocytic Grin2a knockdown or overexpression in the hippocampus, and a cell model of primary cultured hippocampal astrocytes, we reveal a novel mechanism that astrocytic GluN2A alleviates SD-induced increases in Aβ. We demonstrated that astrocytic GluN2A mainly affected Aβ degradation and clearance through regulating degradation enzyme neprilysin and Aquaporin-4 (AQP4), via the calcineurin/NFAT pathway. Our study provides supportive evidence for the novel role and mechanism of astrocytic GluN2A in Aβ elimination, which would contribute to the discovery of new therapeutic strategies for Aβ-related diseases such as AD.
Collapse
Affiliation(s)
- Xiaoqiang Du
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Hongqi Wang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Siyu Liu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Yizhi Song
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Xinyue Chen
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Ziyan Chen
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Ruying Zhou
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Jiahe Du
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Wanning Zhang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Ruiqi Gao
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Hui Li
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Guitao Zhang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Xin Mao
- Department of Radiology, Peking University Third Hospital, Beijing 100068, China.
| | - Lirong Chang
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Yan Wu
- Department of Anatomy, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100069, China; College of Veterinary Medicine, Beijing University of Agriculture, Beijing 100096, China.
| |
Collapse
|
2
|
Park HB, Kim H, Han D. In-Depth Proteome Profiling of the Hippocampus of LDLR Knockout Mice Reveals Alternation in Synaptic Signaling Pathway. Proteomics 2025; 25:e202400152. [PMID: 39548955 DOI: 10.1002/pmic.202400152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/06/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024]
Abstract
The low-density lipoprotein receptor (LDLR) is a major apolipoprotein receptor that regulates cholesterol homeostasis. LDLR deficiency is associated with cognitive impairment by the induction of synaptopathy in the hippocampus. Despite the close relationship between LDLR and neurodegenerative disorders, proteomics research for protein profiling in the LDLR knockout (KO) model remains insufficient. Therefore, understanding LDLR KO-mediated differential protein expression within the hippocampus is crucial for elucidating a role of LDLR in neurodegenerative disorders. In this study, we conducted first-time proteomic profiling of hippocampus tissue from LDLR KO mice using tandem mass tag (TMT)-based MS analysis. LDLR deficiency induces changes in proteins associated with the transport of diverse molecules, and activity of kinase and catalyst within the hippocampus. Additionally, significant alterations in the expression of components in the major synaptic pathways were found. Furthermore, these synaptic effects were verified using a data-independent acquisition (DIA)-based proteomic method. Our data will serve as a valuable resource for further studies to discover the molecular function of LDLR in neurodegenerative disorders.
Collapse
Affiliation(s)
- Hong-Beom Park
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, South Korea
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hyeyoon Kim
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Dohyun Han
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul, South Korea
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
3
|
Adeoye T, Shah SI, Ullah G. Systematic Analysis of Biological Processes Reveals Gene Co-expression Modules Driving Pathway Dysregulation in Alzheimer's Disease. Aging Dis 2024:AD.2024.0429. [PMID: 38913039 DOI: 10.14336/ad.2024.0429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024] Open
Abstract
Alzheimer's disease (AD) manifests as a complex systems pathology with intricate interplay among various genes and biological processes. Traditional differential gene expression (DEG) analysis, while commonly employed to characterize AD-driven perturbations, does not sufficiently capture the full spectrum of underlying biological processes. Utilizing single-nucleus RNA-sequencing data from postmortem brain samples across key regions-middle temporal gyrus, superior frontal gyrus, and entorhinal cortex-we provide a comprehensive systematic analysis of disrupted processes in AD. We go beyond the DEG-centric analysis by integrating pathway activity analysis with weighted gene co-expression patterns to comprehensively map gene interconnectivity, identifying region- and cell-type-specific drivers of biological processes associated with AD. Our analysis reveals profound modular heterogeneity in neurons and glia as well as extensive AD-related functional disruptions. Co-expression networks highlighted the extended involvement of astrocytes and microglia in biological processes beyond neuroinflammation, such as calcium homeostasis, glutamate regulation, lipid metabolism, vesicle-mediated transport, and TOR signaling. We find limited representation of DEGs within dysregulated pathways across neurons and glial cells, suggesting that differential gene expression alone may not adequately represent the disease complexity. Further dissection of inferred gene modules revealed distinct dynamics of hub DEGs in neurons versus glia, suggesting that DEGs exert more impact on neurons compared to glial cells in driving modular dysregulations underlying perturbed biological processes. Interestingly, we observe an overall downregulation of astrocyte and microglia modules across all brain regions in AD, indicating a prevailing trend of functional repression in glial cells across these regions. Notable genes from the CALM and HSP90 families emerged as hub genes across neuronal modules in all brain regions, suggesting conserved roles as drivers of synaptic dysfunction in AD. Our findings demonstrate the importance of an integrated, systems-oriented approach combining pathway and network analysis to comprehensively understand the cell-type-specific roles of genes in AD-related biological processes.
Collapse
|
4
|
Chiricosta L, Minuti A, Gugliandolo A, Salamone S, Pollastro F, Mazzon E, Artimagnella O. Cannabinerol Prevents Endoplasmic Reticulum and Mitochondria Dysfunctions in an In Vitro Model of Alzheimer's Disease: A Network-Based Transcriptomic Analysis. Cells 2024; 13:1012. [PMID: 38920643 PMCID: PMC11201759 DOI: 10.3390/cells13121012] [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: 04/09/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Neurodegenerative disorders are affecting millions of people worldwide, impacting the healthcare system of our society. Among them, Alzheimer's disease (AD) is the most common form of dementia, characterized by severe cognitive impairments. Neuropathological hallmarks of AD are β-amyloid (Aβ) plaques and neurofibrillary tangles, as well as endoplasmic reticulum and mitochondria dysfunctions, which finally lead to apoptosis and neuronal loss. Since, to date, there is no definitive cure, new therapeutic and prevention strategies are of crucial importance. In this scenario, cannabinoids are deeply investigated as promising neuroprotective compounds for AD. In this study, we evaluated the potential neuroprotective role of cannabinerol (CBNR) in an in vitro cellular model of AD via next-generation sequencing. We observed that CBNR pretreatment counteracts the Aβ-induced loss of cell viability of differentiated SH-SY5Y cells. Moreover, a network-based transcriptomic analysis revealed that CBNR restores normal mitochondrial and endoplasmic reticulum functions in the AD model. Specifically, the most important genes regulated by CBNR are related mainly to oxidative phosphorylation (COX6B1, OXA1L, MT-CO2, MT-CO3), protein folding (HSPA5) and degradation (CUL3, FBXW7, UBE2D1), and glucose (G6PC3) and lipid (HSD17B7, ERG28, SCD) metabolism. Therefore, these results suggest that CBNR could be a new neuroprotective agent helpful in the prevention of AD dysfunctions.
Collapse
Affiliation(s)
- Luigi Chiricosta
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Aurelio Minuti
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy; (S.S.); (F.P.)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Osvaldo Artimagnella
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
5
|
Chakraborty P, Hasan G. ER-Ca 2+ stores and the regulation of store-operated Ca 2+ entry in neurons. J Physiol 2024; 602:1463-1474. [PMID: 36691983 DOI: 10.1113/jp283827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Key components of endoplasmic reticulum (ER) Ca2+ release and store-operated Ca2+ entry (SOCE) are likely expressed in all metazoan cells. Due to the complexity of canonical Ca2+ entry mechanisms in neurons, the functional significance of ER-Ca2+ release and SOCE has been difficult to identify and establish. In this review we present evidence of how these two related mechanisms of Ca2+ signalling impact multiple aspects of neuronal physiology and discuss their interaction with the better understood classes of ion channels that are gated by either voltage changes or extracellular ligands in neurons. Given how a small imbalance in Ca2+ homeostasis can have strongly detrimental effects on neurons, leading to cell death, it is essential that neuronal SOCE is carefully regulated. We go on to discuss some mechanisms of SOCE regulation that have been identified in Drosophila and mammalian neurons. These include specific splice variants of stromal interaction molecules, different classes of membrane-interacting proteins and an ER-Ca2+ channel. So far these appear distinct from the mechanisms of SOCE regulation identified in non-excitable cells. Finally, we touch upon the significance of these studies in the context of certain human neurodegenerative diseases.
Collapse
Affiliation(s)
- Pragnya Chakraborty
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
- SASTRA University, Thanjavur, Tamil Nadu, India
| | - Gaiti Hasan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| |
Collapse
|
6
|
Adeoye T, Shah SI, Ullah G. Systematic Analysis of Biological Processes Reveals Gene Co-expression Modules Driving Pathway Dysregulation in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585267. [PMID: 38559218 PMCID: PMC10980062 DOI: 10.1101/2024.03.15.585267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease (AD) manifests as a complex systems pathology with intricate interplay among various genes and biological processes. Traditional differential gene expression (DEG) analysis, while commonly employed to characterize AD-driven perturbations, does not sufficiently capture the full spectrum of underlying biological processes. Utilizing single-nucleus RNA-sequencing data from postmortem brain samples across key regions-middle temporal gyrus, superior frontal gyrus, and entorhinal cortex-we provide a comprehensive systematic analysis of disrupted processes in AD. We go beyond the DEG-centric analysis by integrating pathway activity analysis with weighted gene co-expression patterns to comprehensively map gene interconnectivity, identifying region- and cell-type-specific drivers of biological processes associated with AD. Our analysis reveals profound modular heterogeneity in neurons and glia as well as extensive AD-related functional disruptions. Co-expression networks highlighted the extended involvement of astrocytes and microglia in biological processes beyond neuroinflammation, such as calcium homeostasis, glutamate regulation, lipid metabolism, vesicle-mediated transport, and TOR signaling. We find limited representation of DEGs within dysregulated pathways across neurons and glial cells, indicating that differential gene expression alone may not adequately represent the disease complexity. Further dissection of inferred gene modules revealed distinct dynamics of hub DEGs in neurons versus glia, highlighting the differential impact of DEGs on neurons compared to glial cells in driving modular dysregulations underlying perturbed biological processes. Interestingly, we note an overall downregulation of both astrocyte and microglia modules in AD across all brain regions, suggesting a prevailing trend of functional repression in glial cells across these regions. Notable genes, including those of the CALM and HSP90 family genes emerged as hub genes across neuronal modules in all brain regions, indicating conserved roles as drivers of synaptic dysfunction in AD. Our findings demonstrate the importance of an integrated, systems-oriented approach combining pathway and network analysis for a comprehensive understanding of the cell-type-specific roles of genes in AD-related biological processes.
Collapse
Affiliation(s)
- Temitope Adeoye
- Department of Physics, University of South Florida, Tampa, FL 33620
| | - Syed I Shah
- Department of Physics, University of South Florida, Tampa, FL 33620
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620
| |
Collapse
|
7
|
Romagnolo A, Dematteis G, Scheper M, Luinenburg MJ, Mühlebner A, Van Hecke W, Manfredi M, De Giorgis V, Reano S, Filigheddu N, Bortolotto V, Tapella L, Anink JJ, François L, Dedeurwaerdere S, Mills JD, Genazzani AA, Lim D, Aronica E. Astroglial calcium signaling and homeostasis in tuberous sclerosis complex. Acta Neuropathol 2024; 147:48. [PMID: 38418708 PMCID: PMC10901927 DOI: 10.1007/s00401-024-02711-3] [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: 12/06/2023] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
Tuberous Sclerosis Complex (TSC) is a multisystem genetic disorder characterized by the development of benign tumors in various organs, including the brain, and is often accompanied by epilepsy, neurodevelopmental comorbidities including intellectual disability and autism. A key hallmark of TSC is the hyperactivation of the mechanistic target of rapamycin (mTOR) signaling pathway, which induces alterations in cortical development and metabolic processes in astrocytes, among other cellular functions. These changes could modulate seizure susceptibility, contributing to the progression of epilepsy and its associated comorbidities. Epilepsy is characterized by dysregulation of calcium (Ca2+) channels and intracellular Ca2+ dynamics. These factors contribute to hyperexcitability, disrupted synaptogenesis, and altered synchronization of neuronal networks, all of which contribute to seizure activity. This study investigates the intricate interplay between altered Ca2+ dynamics, mTOR pathway dysregulation, and cellular metabolism in astrocytes. The transcriptional profile of TSC patients revealed significant alterations in pathways associated with cellular respiration, ER and mitochondria, and Ca2+ regulation. TSC astrocytes exhibited lack of responsiveness to various stimuli, compromised oxygen consumption rate and reserve respiratory capacity underscoring their reduced capacity to react to environmental changes or cellular stress. Furthermore, our study revealed significant reduction of store operated calcium entry (SOCE) along with strong decrease of basal mitochondrial Ca2+ concentration and Ca2+ influx in TSC astrocytes. In addition, we observed alteration in mitochondrial membrane potential, characterized by increased depolarization in TSC astrocytes. Lastly, we provide initial evidence of structural abnormalities in mitochondria within TSC patient-derived astrocytes, suggesting a potential link between disrupted Ca2+ signaling and mitochondrial dysfunction. Our findings underscore the complexity of the relationship between Ca2+ signaling, mitochondria dynamics, apoptosis, and mTOR hyperactivation. Further exploration is required to shed light on the pathophysiology of TSC and on TSC associated neuropsychiatric disorders offering further potential avenues for therapeutic development.
Collapse
Affiliation(s)
- Alessia Romagnolo
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Mirte Scheper
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Mark J Luinenburg
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wim Van Hecke
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcello Manfredi
- Center on Autoimmune and Allergic Diseases (CAAD), UPO, Novara, Italy
- Department of Translational Medicine, UPO, Novara, Italy
| | - Veronica De Giorgis
- Center on Autoimmune and Allergic Diseases (CAAD), UPO, Novara, Italy
- Department of Translational Medicine, UPO, Novara, Italy
| | - Simone Reano
- Center on Autoimmune and Allergic Diseases (CAAD), UPO, Novara, Italy
| | | | - Valeria Bortolotto
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Jasper J Anink
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Liesbeth François
- Neurosciences Therapeutic Area, UCB Pharma, Braine-L'Alleud, Belgium
| | | | - James D Mills
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Clinical and Experimental Epilepsy, UCL, London, UK
- Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Novara, Italy
| | - Eleonora Aronica
- Department of (Neuro) Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
8
|
Kaar A, Weir MP, Rae MG. Altered neuronal group 1 metabotropic glutamate receptor- and endoplasmic reticulum-mediated Ca 2+ signaling in two rodent models of Alzheimer's disease. Neurosci Lett 2024; 823:137664. [PMID: 38309326 DOI: 10.1016/j.neulet.2024.137664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 01/15/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Calcium mobilization from the endoplasmic reticulum (ER) induced by, for example, IP3 receptor (IP3R) stimulation, and its subsequent crosstalk with extracellular Ca2+ influx mediated through voltage-gated calcium channels (VGCCs) and neuronal store-operated calcium entry (nSOCE), is essential for normal neuronal signaling and cellular homeostasis. However, several studies suggest that chronic calcium dysregulation may play a key role in the onset and/or progression of neurodegenerative conditions, particularly Alzheimer's disease (AD). Here, using early postnatal hippocampal tissue from two transgenic murine models of AD, we provide further evidence that not only are crucial calcium signaling pathways dysregulated, but also that such dysregulation occurs at very early stages of development. Utilizing epifluorescence calcium imaging, we investigated ER-, nSOCE- and VGCC-mediated calcium signaling in cultured primary hippocampal neurons from two transgenic rodent models of AD: 3xTg-AD mice (PS1M146V/APPSWE/TauP301L) and TgF344-AD rats (APPSWE/PS1ΔE9) between 2 and 9 days old. Our results reveal that, in comparison to control hippocampal neurons, those from 3xTg-AD mice possessed significantly greater basal ER calcium levels, as measured by larger responses to I-mGluR-mediated ER Ca2+ mobilization (amplitude; 4 (0-19) vs 21(12-36) a.u., non-Tg vs 3xTg-AD; median difference (95 % Cl) = 14 a.u. (11-18); p = 0.004)) but reduced nSOCE (15 (4-22) vs 8(5-11) a.u., non-Tg vs 3xTg-AD; median difference (95 % Cl) = -7 a.u. (-3- -10 a.u.); p < 0.0001). Furthermore, unlike non-Tg neurons, where depolarization enhanced the amplitude, duration and area under the curve (A.U.C.) of I-mGluR-evoked ER-mediated calcium signals when compared with basal conditions, this was not apparent in 3xTg-AD neurons. Whilst the amplitude of depolarization-enhanced I-mGluR-evoked ER-mediated calcium signals from both non-Tg F344 and TgF344-AD neurons was significantly enhanced relative to basal conditions, the A.U.C. and duration of responses were enhanced significantly upon depolarization in non-Tg F344, but not in TgF344-AD, neurons. Overall, the nature of basal I-mGluR-mediated calcium responses did not differ significantly between non-Tg F344 and TgF344-AD neurons. In summary, our results characterizing ER- and nSOCE-mediated calcium signaling in neurons demonstrate that ER Ca2+ dyshomeostasis is an early and potentially pathogenic event in familial AD.
Collapse
Affiliation(s)
- Aidan Kaar
- Department of Physiology, School of Medicine, University College Cork, Western Gateway Building, Cork, Ireland
| | - Megan P Weir
- Department of Physiology, School of Medicine, University College Cork, Western Gateway Building, Cork, Ireland
| | - Mark G Rae
- Department of Physiology, School of Medicine, University College Cork, Western Gateway Building, Cork, Ireland.
| |
Collapse
|
9
|
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
|
10
|
Griffin JA, Li X, Lehmler HJ, Holland EB. Predicted versus observed activity of PCB mixtures toward the ryanodine receptor. Neurotoxicology 2024; 100:25-34. [PMID: 38065417 PMCID: PMC10842331 DOI: 10.1016/j.neuro.2023.12.003] [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: 07/12/2023] [Revised: 11/15/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Non-dioxin-like polychlorinated biphenyls (NDL PCBs) alter the activity of the ryanodine receptor (RyR), and this activity is linked to developmental neurotoxicity. Most work to date has focused on the activity of single congeners rather than relevant mixtures. The current study assessed the RyR activity of single congeners or binary, tertiary, and complex PCB mixtures. Observed mixture activity was then compared to the expected activity calculated using the concentration addition (CA) model or a RyR-specific neurotoxic equivalency scheme (rNEQ). The predictions of the CA model were consistent with the observed activity of binary mixtures at the lower portion of the concentration-response curve, supporting the additivity of RyR1 active PCBs. Findings also show that minimally active congeners can compete for the RyR1 binding site, and congeners that do not activate the RyR1 do not interfere with the activity of a full agonist. Complex PCB mixtures that mimic PCB profiles detected in indoor air, fish tissue, and the serum of mothers and children activated the RyR1 and displayed similar efficacy and potency regardless of varying congener profiles. Neither the CA model nor the rNEQ perfectly predicted the observed activity of complex mixtures, but predictions were often within one magnitude of change from the observed response. Importantly, PCB mixtures approximating profiles found in environmental samples or human serum displayed RyR1 activity at concentrations reported in published research. The work presented will aid in the development of risk assessment platforms for NDL PCBs and similar compounds toward RyR1 activation and related neurotoxicity.
Collapse
Affiliation(s)
- Justin A Griffin
- Department of Biological Science, California State University of Long Beach, Long Beach, CA, USA
| | - Xueshu Li
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Erika B Holland
- Department of Biological Science, California State University of Long Beach, Long Beach, CA, USA.
| |
Collapse
|
11
|
Griffioen G. Calcium Dyshomeostasis Drives Pathophysiology and Neuronal Demise in Age-Related Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13243. [PMID: 37686048 PMCID: PMC10487569 DOI: 10.3390/ijms241713243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
This review postulates that age-related neurodegeneration entails inappropriate activation of intrinsic pathways to enable brain plasticity through deregulated calcium (Ca2+) signalling. Ca2+ in the cytosol comprises a versatile signal controlling neuronal cell physiology to accommodate adaptive structural and functional changes of neuronal networks (neuronal plasticity) and, as such, is essential for brain function. Although disease risk factors selectively affect different neuronal cell types across age-related neurodegenerative diseases (NDDs), these appear to have in common the ability to impair the specificity of the Ca2+ signal. As a result, non-specific Ca2+ signalling facilitates the development of intraneuronal pathophysiology shared by age-related NDDs, including mitochondrial dysfunction, elevated reactive oxygen species (ROS) levels, impaired proteostasis, and decreased axonal transport, leading to even more Ca2+ dyshomeostasis. These core pathophysiological processes and elevated cytosolic Ca2+ levels comprise a self-enforcing feedforward cycle inevitably spiralling toward high levels of cytosolic Ca2+. The resultant elevated cytosolic Ca2+ levels ultimately gear otherwise physiological effector pathways underlying plasticity toward neuronal demise. Ageing impacts mitochondrial function indiscriminately of the neuronal cell type and, therefore, contributes to the feedforward cycle of pathophysiology development seen in all age-related NDDs. From this perspective, therapeutic interventions to safely restore Ca2+ homeostasis would mitigate the excessive activation of neuronal destruction pathways and, therefore, are expected to have promising neuroprotective potential.
Collapse
|
12
|
Griffin JA, Li X, Lehmler HJ, Holland EB. Predicted Versus Observed Activity of PCB Mixtures Toward the Ryanodine Receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.554299. [PMID: 37662381 PMCID: PMC10473618 DOI: 10.1101/2023.08.22.554299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Non-dioxin-like polychlorinated biphenyls (NDL PCBs) alter the activity of the ryanodine receptor (RyR), and this activity is linked to developmental neurotoxicity. Most work to date has focused on the activity of single congeners rather than relevant mixtures. The current study assessed the RyR activity of single congeners or binary, tertiary, and complex PCB mixtures. Observed mixture activity was then compared to the expected activity calculated using the concentration addition (CA) model or a RyR-specific neurotoxic equivalency scheme (rNEQ). The predictions of the CA model were consistent with the observed activity of binary mixtures at the lower portion of the concentration-response curve, supporting the additivity of RyR1 active PCBs. Findings also show that minimally active congeners can compete for the RyR1 binding site, and congeners that do not activate the RyR1 do not interfere with the activity of a full agonist. Complex PCB mixtures that mimic PCB profiles detected in indoor air, fish tissue, and the serum of mothers and children activated the RyR1 and displayed similar efficacy and potency regardless of varying congener profiles. Neither the CA model nor the rNEQ perfectly predicted the observed activity of complex mixtures, but predictions were often within one magnitude of change from the observed response. Importantly, PCB mixtures approximating profiles found in environmental samples or human serum displayed RyR1 activity at concentrations reported in published research. The work presented will aid in the development of risk assessment platforms for NDL PCBs, and similar compounds, towards RyR1 activation and related neurotoxicity.
Collapse
Affiliation(s)
- Justin A. Griffin
- Department of Biological Science, California State University of Long Beach, Long Beach California
| | - Xueshu Li
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, Iowa City, Iowa
| | - Hans-Joachim Lehmler
- Department of Occupational and Environmental Health, College of Public Health, University of Iowa, Iowa City, Iowa
| | - Erika B. Holland
- Department of Biological Science, California State University of Long Beach, Long Beach California
| |
Collapse
|
13
|
Dhureja M, Arthur R, Soni D, Upadhayay S, Temgire P, Kumar P. Calcium channelopathies in neurodegenerative disorder: an untold story of RyR and SERCA. Expert Opin Ther Targets 2023; 27:1159-1172. [PMID: 37971192 DOI: 10.1080/14728222.2023.2277863] [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: 08/26/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Recent neuroscience breakthroughs have shed light on the sophisticated relationship between calcium channelopathies and movement disorders, exposing a previously undiscovered tale focusing on the Ryanodine Receptor (RyR) and the Sarco/Endoplasmic Reticulum Calcium ATPase (SERCA). Calcium signaling mainly orchestrates neural communication, which regulates synaptic transmission and total network activity. It has been determined that RyR play a significant role in managing neuronal functions, most notably in releasing intracellular calcium from the endoplasmic reticulum. AREAS COVERED It highlights the involvement of calcium channels such as RyR and SERCA in physiological and pathophysiological conditions. EXPERT OPINION Links between RyR and SERCA activity dysregulation, aberrant calcium levels, motor and cognitive dysfunction have brought attention to the importance of RyR and SERCA modulation in neurodegenerative disorders. Understanding the obscure function of these proteins will open up new therapeutic possibilities to address the underlying causes of neurodegenerative diseases. The unreported RyR and SERCA narrative broadens the understanding of calcium channelopathies in movement disorders and calls for more research into cutting-edge therapeutic approaches.
Collapse
Affiliation(s)
- Maanvi Dhureja
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Richmond Arthur
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Divya Soni
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Shubham Upadhayay
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Pooja Temgire
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
14
|
Garcia-Casas P, Rossini M, Filadi R, Pizzo P. Mitochondrial Ca 2+ signaling and Alzheimer's disease: Too much or too little? Cell Calcium 2023; 113:102757. [PMID: 37192560 DOI: 10.1016/j.ceca.2023.102757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, caused by poorly known pathogenic mechanisms and aggravated by delayed therapeutic intervention, that still lacks an effective cure. However, it is clear that some important neurophysiological processes are altered years before the onset of clinical symptoms, offering the possibility of identifying biological targets useful for implementation of new therapies. Of note, evidence has been provided suggesting that mitochondria, pivotal organelles in sustaining neuronal energy demand and modulating synaptic activity, are dysfunctional in AD samples. In particular, alterations in mitochondrial Ca2+ signaling have been proposed as causal events for neurodegeneration, although the exact outcomes and molecular mechanisms of these defects, as well as their longitudinal progression, are not always clear. Here, we discuss the importance of a correct mitochondrial Ca2+ handling for neuronal physiology and summarize the latest findings on dysfunctional mitochondrial Ca2+ pathways in AD, analysing possible consequences contributing to the neurodegeneration that characterizes the disease.
Collapse
Affiliation(s)
- Paloma Garcia-Casas
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy; Department of Biochemistry and Molecular Biology and Physiology, School of Medicine, University of Valladolid, 47003 Valladolid, Spain
| | - Michela Rossini
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy; Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy.
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy; Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy; Study Centre for Neurodegeneration (CESNE), University of Padova, 35131 Padua, Italy.
| |
Collapse
|
15
|
Kumar H, Kulkarni G, Diwan V, Sharma B. Shielding Effect of Ryanodine Receptor Modulator in Rat Model of Autism. Basic Clin Neurosci 2023; 14:247-261. [PMID: 38107532 PMCID: PMC10719969 DOI: 10.32598/bcn.2021.2966.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 05/02/2021] [Accepted: 06/13/2021] [Indexed: 12/19/2023] Open
Abstract
Introduction A neurodevelopmental disorder, autism is typically identified with three primary behavioral consequences, such as social impairment, communication problems, and limited or stereotypical behavior. Because of its co-morbidity and lack of therapeutic options, autism is a global economic burden. A short chain of fatty acid, propionic acid is formed biologically by the gut microbiome. Propionic acid levels that are too high can cause leaky intestines, which can lead to autism-like symptoms. Methods To induce autism, male Albino Wistar rats were given propionic acid (250 mg/kg/po on the 21st, 22nd, and 23rd postnatal days). Rats also received a ryanodine receptor antagonist (Ruthenium red: 3 mg/kg/po; postnatal 21st to 50th day) to see what influence it had on propionic acid-induced autism. Anxiety, social behavior, and repeated behaviors were all assessed, as well as oxidative stress, inflammatory indicators, neuro signaling proteins, and blood-brain barrier permeability. Results Ruthenium red was found to counter the propionic acid-induced increases in anxiety, repetitive behavior prefrontal cortex levels of IL-6, TNF-α, TBARS, Evans blue leakage, and water content along with decreases in social behavior, IL-10, and GSH followed by hippocampus CREB and BDNF levels. Conclusion Ryanodine receptor antagonists presented a neuroprotective effect in propionic acid-induced conditions like autism by modulatory effects on social and repetitive behavior, oxidative stress, neuroinflammation, and neuroprotein changes. Ryanodine receptors can be further explored in depth to manage autism as a condition. Highlights Ruthenium red can reduce the propionic acid-induced anxiety of rats with autism.Ruthenium red can improve the propionic acid-induced changes in repetitive behavior of rats with autism.Ruthenium red can reduce the propionic acid-induced social behavior dysfunction in rats with autism. Plain Language Summary Autism is a complex heterogeneous neurodevelopmental disorder mainly diagnosed with social behavior dysfunction, communication problems, and repetitive behavior. Due to high comorbidity and multiple unknown factors involvement, its exact etiology remains unclear, and so no successful treatment is available. Among the environmentally produced models of autism in rats, the most common is created by propionic acid (PPA). With short-chain type fatty acid, PPA is one of the mediators for the cycle of cell metabolism. This study attempted to study the effect of a ryano-dine receptor antagonist (Ruthenium red) on PPA-induced Anxiety, social behavior dysfunction, and repeated behaviors in rats with autism. The results showed the modulatory effects of Ruthenium red PPA-induced conditions including social and repetitive behavior, oxidative stress, neuroinflammation, and neuroprotein changes in rats with autism.
Collapse
Affiliation(s)
- Hariom Kumar
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Nodia, India
| | - G.T. Kulkarni
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Nodia, India
| | - Vishal Diwan
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Australia
| | - Bhupesh Sharma
- Department of Pharmacology, Amity Institute of Pharmacy, Amity University, Nodia, India
| |
Collapse
|
16
|
Knutson KR, Whiteman ST, Alcaino C, Mercado-Perez A, Finholm I, Serlin HK, Bellampalli SS, Linden DR, Farrugia G, Beyder A. Intestinal enteroendocrine cells rely on ryanodine and IP 3 calcium store receptors for mechanotransduction. J Physiol 2023; 601:287-305. [PMID: 36428286 PMCID: PMC9840706 DOI: 10.1113/jp283383] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/24/2022] [Indexed: 11/27/2022] Open
Abstract
Enteroendocrine cells (EECs) are specialized sensors of luminal forces and chemicals in the gastrointestinal (GI) epithelium that respond to stimulation with a release of signalling molecules such as serotonin (5-HT). For mechanosensitive EECs, force activates Piezo2 channels, which generate a very rapidly activating and inactivating (∼10 ms) cationic (Na+ , K+ , Ca2+ ) receptor current. Piezo2 receptor currents lead to a large and persistent increase in intracellular calcium (Ca2+ ) that lasts many seconds to sometimes minutes, suggesting signal amplification. However, intracellular calcium dynamics in EEC mechanotransduction remain poorly understood. The aim of this study was to determine the role of Ca2+ stores in EEC mechanotransduction. Mechanical stimulation of a human EEC cell model (QGP-1) resulted in a rapid increase in cytoplasmic Ca2+ and a slower decrease in ER stores Ca2+ , suggesting the involvement of intracellular Ca2+ stores. Comparing murine primary colonic EECs with colonocytes showed expression of intercellular Ca2+ store receptors, a similar expression of IP3 receptors, but a >30-fold enriched expression of Ryr3 in EECs. In mechanically stimulated primary EECs, Ca2+ responses decreased dramatically by emptying stores and pharmacologically blocking IP3 and RyR1/3 receptors. RyR3 genetic knockdown by siRNA led to a significant decrease in mechanosensitive Ca2+ responses and 5-HT release. In tissue, pressure-induced increase in the Ussing short circuit current was significantly decreased by ryanodine receptor blockade. Our data show that mechanosensitive EECs use intracellular Ca2+ stores to amplify mechanically induced Ca2+ entry, with RyR3 receptors selectively expressed in EECs and involved in Ca2+ signalling, 5-HT release and epithelial secretion. KEY POINTS: A population of enteroendocrine cells (EECs) are specialized mechanosensors of the gastrointestinal (GI) epithelium that respond to mechanical stimulation with the release of important signalling molecules such as serotonin. Mechanical activation of these EECs leads to an increase in intracellular calcium (Ca2+ ) with a longer duration than the stimulus, suggesting intracellular Ca2+ signal amplification. In this study, we profiled the expression of intracellular Ca2+ store receptors and found an enriched expression of the intracellular Ca2+ receptor Ryr3, which contributed to the mechanically evoked increases in intracellular calcium, 5-HT release and epithelial secretion. Our data suggest that mechanosensitive EECs rely on intracellular Ca2+ stores and are selective in their use of Ryr3 for amplification of intracellular Ca2+ . This work advances our understanding of EEC mechanotransduction and may provide novel diagnostic and therapeutic targets for GI motility disorders.
Collapse
Affiliation(s)
- Kaitlyn R. Knutson
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Sara T. Whiteman
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Constanza Alcaino
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arnaldo Mercado-Perez
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Medical Scientist Training Program (MSTP), Mayo Clinic, Rochester, Minnesota
| | - Isabelle Finholm
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Hannah K. Serlin
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Shreya S. Bellampalli
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Medical Scientist Training Program (MSTP), Mayo Clinic, Rochester, Minnesota
| | - David R. Linden
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Division of Gastroenterology &Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, Minnesota
- Division of Gastroenterology &Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
17
|
Wang ZW, Niu L, Riaz S. Regulation of Ryanodine Receptor-Dependent Neurotransmitter Release by AIP, Calstabins, and Presenilins. ADVANCES IN NEUROBIOLOGY 2023; 33:287-304. [PMID: 37615871 DOI: 10.1007/978-3-031-34229-5_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Ryanodine receptors (RyRs) are Ca2+ release channels located in the endoplasmic reticulum membrane. Presynaptic RyRs play important roles in neurotransmitter release and synaptic plasticity. Recent studies suggest that the proper function of presynaptic RyRs relies on several regulatory proteins, including aryl hydrocarbon receptor-interacting protein, calstabins, and presenilins. Dysfunctions of these regulatory proteins can greatly impact neurotransmitter release and synaptic plasticity by altering the function or expression of RyRs. This chapter aims to describe the interaction between these proteins and RyRs, elucidating their crucial role in regulating synaptic function.
Collapse
Affiliation(s)
- Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Longgang Niu
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Sadaf Riaz
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
18
|
Wang ZW, Riaz S, Niu L. Roles and Sources of Calcium in Synaptic Exocytosis. ADVANCES IN NEUROBIOLOGY 2023; 33:139-170. [PMID: 37615866 DOI: 10.1007/978-3-031-34229-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Calcium ions (Ca2+) play a critical role in triggering neurotransmitter release. The rate of release is directly related to the concentration of Ca2+ at the presynaptic site, with a supralinear relationship. There are two main sources of Ca2+ that trigger synaptic vesicle fusion: influx through voltage-gated Ca2+ channels in the plasma membrane and release from the endoplasmic reticulum via ryanodine receptors. This chapter will cover the sources of Ca2+ at the presynaptic nerve terminal, the relationship between neurotransmitter release rate and Ca2+ concentration, and the mechanisms that achieve the necessary Ca2+ concentrations for triggering synaptic exocytosis at the presynaptic site.
Collapse
Affiliation(s)
- Zhao-Wen Wang
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Sadaf Riaz
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Longgang Niu
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
19
|
Mustaly-Kalimi S, Gallegos W, Marr RA, Gilman-Sachs A, Peterson DA, Sekler I, Stutzmann GE. Protein mishandling and impaired lysosomal proteolysis generated through calcium dysregulation in Alzheimer's disease. Proc Natl Acad Sci U S A 2022; 119:e2211999119. [PMID: 36442130 PMCID: PMC9894236 DOI: 10.1073/pnas.2211999119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022] Open
Abstract
Impairments in neural lysosomal- and autophagic-mediated degradation of cellular debris contribute to neuritic dystrophy and synaptic loss. While these are well-characterized features of neurodegenerative disorders such as Alzheimer's disease (AD), the upstream cellular processes driving deficits in pathogenic protein mishandling are less understood. Using a series of fluorescent biosensors and optical imaging in model cells, AD mouse models and human neurons derived from AD patients, we reveal a previously undescribed cellular signaling cascade underlying protein mishandling mediated by intracellular calcium dysregulation, an early component of AD pathogenesis. Increased Ca2+ release via the endoplasmic reticulum (ER)-resident ryanodine receptor (RyR) is associated with reduced expression of the lysosome proton pump vacuolar-ATPase (vATPase) subunits (V1B2 and V0a1), resulting in lysosome deacidification and disrupted proteolytic activity in AD mouse models and human-induced neurons (HiN). As a result of impaired lysosome digestive capacity, mature autophagosomes with hyperphosphorylated tau accumulated in AD murine neurons and AD HiN, exacerbating proteinopathy. Normalizing AD-associated aberrant RyR-Ca2+ signaling with the negative allosteric modulator, dantrolene (Ryanodex), restored vATPase levels, lysosomal acidification and proteolytic activity, and autophagic clearance of intracellular protein aggregates in AD neurons. These results highlight that prior to overt AD histopathology or cognitive deficits, aberrant upstream Ca2+ signaling disrupts lysosomal acidification and contributes to pathological accumulation of intracellular protein aggregates. Importantly, this is demonstrated in animal models of AD, and in human iPSC-derived neurons from AD patients. Furthermore, pharmacological suppression of RyR-Ca2+ release rescued proteolytic function, revealing a target for therapeutic intervention that has demonstrated effects in clinically-relevant assays.
Collapse
Affiliation(s)
- Sarah Mustaly-Kalimi
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Wacey Gallegos
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Robert A. Marr
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Alice Gilman-Sachs
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University of Medicine and Science, Immunology, and Infection, North Chicago, IL60064
| | - Daniel A. Peterson
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Science and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva84105, Israel
| | - Grace E. Stutzmann
- Center for Neurodegenerative Disease and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| |
Collapse
|
20
|
Cappabianca L, Sebastiano M, Ruggieri M, Sbaffone M, Zelli V, Farina AR, Mackay AR. Doxorubicin-Induced TrkAIII Activation: A Selection Mechanism for Resistant Dormant Neuroblastoma Cells. Int J Mol Sci 2022; 23:ijms231810895. [PMID: 36142807 PMCID: PMC9503591 DOI: 10.3390/ijms231810895] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Patients with advanced neuroblastoma (NB) receive multimodal clinical therapy, including the potent anthracycline chemotherapy drug doxorubicin (Dox). The acquisition of Dox resistance, however, is a major barrier to a sustained response and leads to a poor prognosis in advanced disease states, reinforcing the need to identify and inhibit Dox resistance mechanisms. In this context, we report on the identification and inhibition of a novel Dox resistance mechanism. This mechanism is characterized by the Dox-induced activation of the oncogenic TrkAIII alternative splice variant, resulting in increased Dox resistance, and is blocked by lestaurtinib, entrectinib, and crizotinib tyrosine kinase and LY294002 IP3-K inhibitors. Using time lapse live cell imaging, conventional and co-immunoprecipitation Western blots, RT-PCR, and inhibitor studies, we report that the Dox-induced TrkAIII activation correlates with proliferation inhibition and is CDK1- and Ca2+-uniporter-independent. It is mediated by ryanodine receptors; involves Ca2+-dependent interactions between TrkAIII, calmodulin and Hsp90; requires oxygen and oxidation; occurs within assembled ERGICs; and does not occur with fully spliced TrkA. The inhibitory effects of lestaurtinib, entrectinib, crizotinib, and LY294002 on the Dox-induced TrkAIII and Akt phosphorylation and resistance confirm roles for TrkAIII and IP3-K consistent with Dox-induced, TrkAIII-mediated pro-survival IP3K/Akt signaling. This mechanism has the potential to select resistant dormant TrkAIII-expressing NB cells, supporting the use of Trk inhibitors during Dox therapy in TrkAIII-expressing NBs.
Collapse
|
21
|
Yu Y, Li W, Jiang J. TRPC channels as emerging targets for seizure disorders. Trends Pharmacol Sci 2022; 43:787-798. [PMID: 35840362 PMCID: PMC9378536 DOI: 10.1016/j.tips.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/09/2022] [Accepted: 06/17/2022] [Indexed: 10/17/2022]
Abstract
Epilepsy is characterized by seizures of diverse types that affect about 1-2% of the population worldwide. Current antiseizure medications are unsatisfactory, as they merely provide symptomatic relief, are ineffective in about one-third of patients, and cause unbearable adverse effects. Transient receptor potential canonical (TRPC) channels are a group of nonselective cation channels involved in many physiological functions. In this review, we provide an overview of recent preclinical studies using both genetic and pharmacological strategies that reveal these receptor-operated calcium-permeable channels may also play fundamental roles in many aspects of epileptic seizures. We also propose that TRPC channels represent appealing targets for epilepsy treatment, with a goal of helping to advance the discovery and development of new antiseizure and/or antiepileptogenic therapies.
Collapse
Affiliation(s)
- Ying Yu
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences and Drug Discovery Center, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
22
|
Role of Microglia and Astrocytes in Alzheimer’s Disease: From Neuroinflammation to Ca2+ Homeostasis Dysregulation. Cells 2022; 11:cells11172728. [PMID: 36078138 PMCID: PMC9454513 DOI: 10.3390/cells11172728] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia worldwide, with a complex, poorly understood pathogenesis. Cerebral atrophy, amyloid-β (Aβ) plaques, and neurofibrillary tangles represent the main pathological hallmarks of the AD brain. Recently, neuroinflammation has been recognized as a prominent feature of the AD brain and substantial evidence suggests that the inflammatory response modulates disease progression. Additionally, dysregulation of calcium (Ca2+) homeostasis represents another early factor involved in the AD pathogenesis, as intracellular Ca2+ concentration is essential to ensure proper cellular and neuronal functions. Although growing evidence supports the involvement of Ca2+ in the mechanisms of neurodegeneration-related inflammatory processes, scant data are available on its contribution in microglia and astrocytes functioning, both in health and throughout the AD continuum. Nevertheless, AD-related aberrant Ca2+ signalling in astrocytes and microglia is crucially involved in the mechanisms underpinning neuroinflammatory processes that, in turn, impact neuronal Ca2+ homeostasis and brain function. In this light, we attempted to provide an overview of the current understanding of the interactions between the glia cells-mediated inflammatory responses and the molecular mechanisms involved in Ca2+ homeostasis dysregulation in AD.
Collapse
|
23
|
Adeoye T, Shah SI, Demuro A, Rabson DA, Ullah G. Upregulated Ca 2+ Release from the Endoplasmic Reticulum Leads to Impaired Presynaptic Function in Familial Alzheimer's Disease. Cells 2022; 11:2167. [PMID: 35883609 PMCID: PMC9315668 DOI: 10.3390/cells11142167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/28/2022] [Accepted: 07/02/2022] [Indexed: 12/10/2022] Open
Abstract
Neurotransmitter release from presynaptic terminals is primarily regulated by rapid Ca2+ influx through membrane-resident voltage-gated Ca2+ channels (VGCCs). Moreover, accumulating evidence indicates that the endoplasmic reticulum (ER) is extensively present in axonal terminals of neurons and plays a modulatory role in synaptic transmission by regulating Ca2+ levels. Familial Alzheimer's disease (FAD) is marked by enhanced Ca2+ release from the ER and downregulation of Ca2+ buffering proteins. However, the precise consequence of impaired Ca2+ signaling within the vicinity of VGCCs (active zone (AZ)) on exocytosis is poorly understood. Here, we perform in silico experiments of intracellular Ca2+ signaling and exocytosis in a detailed biophysical model of hippocampal synapses to investigate the effect of aberrant Ca2+ signaling on neurotransmitter release in FAD. Our model predicts that enhanced Ca2+ release from the ER increases the probability of neurotransmitter release in FAD. Moreover, over very short timescales (30-60 ms), the model exhibits activity-dependent and enhanced short-term plasticity in FAD, indicating neuronal hyperactivity-a hallmark of the disease. Similar to previous observations in AD animal models, our model reveals that during prolonged stimulation (~450 ms), pathological Ca2+ signaling increases depression and desynchronization with stimulus, causing affected synapses to operate unreliably. Overall, our work provides direct evidence in support of a crucial role played by altered Ca2+ homeostasis mediated by intracellular stores in FAD.
Collapse
Affiliation(s)
- Temitope Adeoye
- Department of Physics, University of South Florida, Tampa, FL 33620, USA; (T.A.); (S.I.S.); (D.A.R.)
| | - Syed I. Shah
- Department of Physics, University of South Florida, Tampa, FL 33620, USA; (T.A.); (S.I.S.); (D.A.R.)
| | - Angelo Demuro
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA;
| | - David A. Rabson
- Department of Physics, University of South Florida, Tampa, FL 33620, USA; (T.A.); (S.I.S.); (D.A.R.)
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620, USA; (T.A.); (S.I.S.); (D.A.R.)
| |
Collapse
|
24
|
Rebbeck R, Ginsburg KS, Ko CY, Fasoli A, Rusch K, Cai GF, Dong X, Thomas DD, Bers DM, Cornea RL. Synergistic FRET assays for drug discovery targeting RyR2 channels. J Mol Cell Cardiol 2022; 168:13-23. [PMID: 35405106 PMCID: PMC10088286 DOI: 10.1016/j.yjmcc.2022.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/09/2022] [Accepted: 04/05/2022] [Indexed: 10/18/2022]
Abstract
A key therapeutic target for heart failure and arrhythmia is the deleterious leak through sarcoplasmic reticulum (SR) ryanodine receptor 2 (RyR2) calcium release channels. We have previously developed methods to detect the pathologically leaky state of RyR2 in adult cardiomyocytes by monitoring RyR2 binding to either calmodulin (CaM) or a biosensor peptide (DPc10). Here, we test whether these complementary binding measurements are effective as high-throughput screening (HTS) assays to discover small molecules that target leaky RyR2. Using FRET, we developed and validated HTS procedures under conditions that mimic a pathological state, to screen the library of 1280 pharmaceutically active compounds (LOPAC) for modulators of RyR2 in cardiac SR membrane preparations. Complementary FRET assays with acceptor-labeled CaM and DPc10 were used for Hit prioritization based on the opposing binding properties of CaM vs. DPc10. This approach narrowed the Hit list to one compound, Ro 90-7501, which altered FRET to suggest increased RyR2-CaM binding and decreased DPc10 binding. Follow-up studies revealed that Ro 90-7501 does not detrimentally affect myocyte Ca2+ transients. Moreover, Ro 90-7501 partially inhibits overall Ca2+ leak, as assessed by Ca2+ sparks in permeabilized rat cardiomyocytes. Together, these results demonstrate (1) the effectiveness of our HTS approach where two complementary assays synergize for Hit ranking and (2) a drug discovery process that combines high-throughput, high-precision in vitro structural assays with in situ myocyte assays of the pathologic RyR2 leak. These provide a drug discovery platform compatible with large-scale HTS campaigns, to identify agents that inhibit RyR2 for therapeutic development.
Collapse
Affiliation(s)
- RobynT Rebbeck
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, MN, USA
| | | | - Christopher Y Ko
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Anna Fasoli
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Katherine Rusch
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, MN, USA
| | - George F Cai
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, MN, USA
| | - Xiaoqiong Dong
- Department of Pharmacology, University of California, Davis, CA, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, MN, USA; Photonic Pharma LLC, Minneapolis, MN, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, MN, USA; Photonic Pharma LLC, Minneapolis, MN, USA.
| |
Collapse
|
25
|
Paschou M, Papazafiri P, Charalampous C, Zachariadis M, Dedos SG, Doxakis E. Neuronal microRNAs safeguard ER Ca 2+ homeostasis and attenuate the unfolded protein response upon stress. Cell Mol Life Sci 2022; 79:373. [PMID: 35727337 PMCID: PMC11073139 DOI: 10.1007/s00018-022-04398-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/23/2022] [Accepted: 05/21/2022] [Indexed: 11/30/2022]
Abstract
Ca2+ is a critical mediator of neurotransmitter release, synaptic plasticity, and gene expression, but also excitotoxicity. Ca2+ signaling and homeostasis are coordinated by an intricate network of channels, pumps, and calcium-binding proteins, which must be rapidly regulated at all expression levels. Τhe role of neuronal miRNAs in regulating ryanodine receptors (RyRs) and inositol 1,4,5-triphosphate receptors (IP3Rs) was investigated to understand the underlying mechanisms that modulate ER Ca2+ release. RyRs and IP3Rs are critical in mounting and propagating cytosolic Ca2+ signals by functionally linking the ER Ca2+ content, while excessive ER Ca2+ release via these receptors is central to the pathophysiology of a wide range of neurological diseases. Herein, two brain-restricted microRNAs, miR-124-3p and miR-153-3p, were found to bind to RyR1-3 and IP3R3 3'UTRs, and suppress their expression at both the mRNA and protein level. Ca2+ imaging studies revealed that overexpression of these miRNAs reduced ER Ca2+ release upon RyR/IP3R activation, but had no effect on [Ca2+]i under resting conditions. Interestingly, treatments that cause excessive ER Ca2+ release decreased expression of these miRNAs and increased expression of their target ER Ca2+ channels, indicating interdependence of miRNAs, RyRs, and IP3Rs in Ca2+ homeostasis. Furthermore, by maintaining the ER Ca2+ content, miR-124 and miR-153 reduced cytosolic Ca2+ overload and preserved protein-folding capacity by attenuating PERK signaling. Overall, this study shows that miR-124-3p and miR-153-3p fine-tune ER Ca2+ homeostasis and alleviate ER stress responses.
Collapse
Affiliation(s)
- Maria Paschou
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Soranou Efesiou 4, 11527, Athens, Greece
- Department of Biology, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, 15784, Athens, Greece
| | - Panagiota Papazafiri
- Department of Biology, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, 15784, Athens, Greece
| | - Chrysanthi Charalampous
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Soranou Efesiou 4, 11527, Athens, Greece
| | - Michael Zachariadis
- Department of Biology, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, 15784, Athens, Greece
- Material and Chemical Characterization Facility (MC2), Faculty of Science, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Skarlatos G Dedos
- Department of Biology, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, 15784, Athens, Greece.
| | - Epaminondas Doxakis
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Soranou Efesiou 4, 11527, Athens, Greece.
| |
Collapse
|
26
|
Chou YH, Sundman M, Ton That V, Green J, Trapani C. Cortical excitability and plasticity in Alzheimer's disease and mild cognitive impairment: A systematic review and meta-analysis of transcranial magnetic stimulation studies. Ageing Res Rev 2022; 79:101660. [PMID: 35680080 DOI: 10.1016/j.arr.2022.101660] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 05/13/2022] [Accepted: 05/30/2022] [Indexed: 11/01/2022]
Abstract
BACKGROUND Transcranial magnetic stimulation (TMS) is a non-invasive neuromodulation technique. When stimulation is applied over the primary motor cortex and coupled with electromyography measures, TMS can probe functions of cortical excitability and plasticity in vivo. The purpose of this meta-analysis is to evaluate the utility of TMS-derived measures for differentiating patients with Alzheimer's disease (AD) and mild cognitive impairment (MCI) from cognitively normal older adults (CN). METHODS Databases searched included PubMed, Embase, APA PsycInfo, Medline, and CINAHL Plus from inception to July 2021. RESULTS Sixty-one studies with a total of 2728 participants (1454 patients with AD, 163 patients with MCI, and 1111 CN) were included. Patients with AD showed significantly higher cortical excitability, lower cortical inhibition, and impaired cortical plasticity compared to the CN cohorts. Patients with MCI exhibited increased cortical excitability and reduced plasticity compared to the CN cohort. Additionally, lower cognitive performance was significantly associated with higher cortical excitability and lower inhibition. No seizure events due to TMS were reported, and the mild adverse response rate is approximately 3/1000 (i.e., 9/2728). CONCLUSIONS Findings of our meta-analysis demonstrate the potential of using TMS-derived cortical excitability and plasticity measures as diagnostic biomarkers and therapeutic targets for AD and MCI.
Collapse
Affiliation(s)
- Ying-Hui Chou
- Brain Imaging and TMS Laboratory, Department of Psychology, University of Arizona, Tucson, USA; Evelyn F McKnight Brain Institute, Arizona Center on Aging, and BIO5 Institute, University of Arizona, Tucson, USA.
| | - Mark Sundman
- Brain Imaging and TMS Laboratory, Department of Psychology, University of Arizona, Tucson, USA
| | - Viet Ton That
- Brain Imaging and TMS Laboratory, Department of Psychology, University of Arizona, Tucson, USA
| | - Jacob Green
- Brain Imaging and TMS Laboratory, Department of Psychology, University of Arizona, Tucson, USA
| | - Chrisopher Trapani
- Brain Imaging and TMS Laboratory, Department of Psychology, University of Arizona, Tucson, USA
| |
Collapse
|
27
|
Jyoti Dutta B, Singh S, Seksaria S, Das Gupta G, Bodakhe SH, Singh A. Potential role of IP3/Ca 2+ signaling and phosphodiesterases: Relevance to neurodegeneration in Alzheimer's disease and possible therapeutic strategies. Biochem Pharmacol 2022; 201:115071. [PMID: 35525328 DOI: 10.1016/j.bcp.2022.115071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/02/2022]
Abstract
Despite large investments by industry and governments, no disease-modifying medications for the treatment of patients with Alzheimer's disease (AD) have been found. The failures of various clinical trials indicate the need for a more in-depth understanding of the pathophysiology of AD and for innovative therapeutic strategies for its treatment. Here, we review the rational for targeting IP3 signaling, cytosolic calcium dysregulation, phosphodiesterases (PDEs), and secondary messengers like cGMP and cAMP, as well as their correlations with the pathophysiology of AD. Various drugs targeting these signaling cascades are still in pre-clinical and clinical trials which support the ideas presented in this article. Further, we describe different molecular mechanisms and medications currently being used in various pre-clinical and clinical trials involving IP3/Ca+2 signaling. We also highlight various isoforms, as well as the functions and pharmacology of the PDEs broadly expressed in different parts of the brain and attempt to unravel the potential benefits of PDE inhibitors for use as novel medications to alleviate the pathogenesis of AD.
Collapse
Affiliation(s)
- Bhaskar Jyoti Dutta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Sanket Seksaria
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur - 495009, Chhattisgarh, India
| | - Amrita Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India.
| |
Collapse
|
28
|
Inflammation and Nitro-oxidative Stress as Drivers of Endocannabinoid System Aberrations in Mood Disorders and Schizophrenia. Mol Neurobiol 2022; 59:3485-3503. [PMID: 35347586 DOI: 10.1007/s12035-022-02800-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/13/2022] [Indexed: 01/02/2023]
Abstract
The endocannabinoid system (ECS) is composed of the endocannabinoid ligands anandamide (AEA) and 2-arachidonoylgycerol (2-AG), their target cannabinoid receptors (CB1 and CB2) and the enzymes involved in their synthesis and metabolism (N-acyltransferase and fatty acid amide hydrolase (FAAH) in the case of AEA and diacylglycerol lipase (DAGL) and monoacylglycerol lipase (MAGL) in the case of 2-AG). The origins of ECS dysfunction in major neuropsychiatric disorders remain to be determined, and this paper explores the possibility that they may be associated with chronically increased nitro-oxidative stress and activated immune-inflammatory pathways, and it examines the mechanisms which might be involved. Inflammation and nitro-oxidative stress are associated with both increased CB1 expression, via increased activity of the NADPH oxidases NOX4 and NOX1, and increased CNR1 expression and DNA methylation; and CB2 upregulation via increased pro-inflammatory cytokine levels, binding of the transcription factor Nrf2 to an antioxidant response element in the CNR2 promoter region and the action of miR-139. CB1 and CB2 have antagonistic effects on redox signalling, which may result from a miRNA-enabled negative feedback loop. The effects of inflammation and oxidative stress are detailed in respect of AEA and 2-AG levels, via effects on calcium homeostasis and phospholipase A2 activity; on FAAH activity, via nitrosylation/nitration of functional cysteine and/or tyrosine residues; and on 2-AG activity via effects on MGLL expression and MAGL. Finally, based on these detailed molecular neurobiological mechanisms, it is suggested that cannabidiol and dimethyl fumarate may have therapeutic potential for major depressive disorder, bipolar disorder and schizophrenia.
Collapse
|
29
|
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: 0.3] [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
|
30
|
Identification of a dihydropyridine scaffold that blocks ryanodine receptors. iScience 2022; 25:103706. [PMID: 35059610 PMCID: PMC8760560 DOI: 10.1016/j.isci.2021.103706] [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] [Received: 09/09/2021] [Revised: 11/16/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022] Open
Abstract
Ryanodine receptors (RyRs) are large, intracellular ion channels that control Ca2+ release from the sarco/endoplasmic reticulum. Dysregulation of RyRs in skeletal muscle, heart, and brain has been implicated in various muscle pathologies, arrhythmia, heart failure, and Alzheimer's disease. Therefore, there is considerable interest in therapeutically targeting RyRs to normalize Ca2+ homeostasis in scenarios involving RyR dysfunction. Here, a simple invertebrate screening platform was used to discover new chemotypes targeting RyRs. The approach measured Ca2+ signals evoked by cyclic adenosine 5′-diphosphate ribose, a second messenger that sensitizes RyRs. From a 1,534-compound screen, FLI-06 (currently described as a Notch “inhibitor”) was identified as a potent blocker of RyR activity. Two closely related tyrosine kinase inhibitors that stimulate and inhibit Ca2+ release through RyRs were also resolved. Therefore, this simple screen yielded RyR scaffolds tractable for development and revealed an unexpected linkage between RyRs and trafficking events in the early secretory pathway. FLI-06 inhibits transport in the secretory pathway via an unknown mechanism An invertebrate screening platform revealed FLI-06 blocks intracellular Ca2+ release FLI-06 acts as a potent, cell-permeable ryanodine receptor (RyR) blocker The para-substituted dihydropyridine chemotype is a new scaffold for RyR modulation
Collapse
|
31
|
Chavda V, Singh K, Patel V, Mishra M, Mishra AK. Neuronal Glial Crosstalk: Specific and Shared Mechanisms in Alzheimer’s Disease. Brain Sci 2022; 12:brainsci12010075. [PMID: 35053818 PMCID: PMC8773743 DOI: 10.3390/brainsci12010075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023] Open
Abstract
The human brain maintains billions of neurons functional across the lifespan of the individual. The glial, supportive cells of the brain are indispensable to neuron elasticity. They undergo various states (active, reactive, macrophage, primed, resting) and carefully impose either quick repair or the cleaning of injured neurons to avoid damage extension. Identifying the failure of these interactions involving the relation of the input of glial cells to the inception and/or progression of chronic neurodegenerative diseases (ND) is crucial in identifying therapeutic options, given the well-built neuro-immune module of these diseases. In the present review, we scrutinize different interactions and important factors including direct cell–cell contact, intervention by the CD200 system, various receptors present on their surfaces, CXC3RI and TREM2, and chemokines and cytokines with special reference to Alzheimer’s disease (AD). The present review of the available literature will elucidate the contribution of microglia and astrocytes to the pathophysiology of AD, thus evidencing glial cells as obligatory transducers of pathology and superlative targets for interference.
Collapse
Affiliation(s)
- Vishal Chavda
- Division of Anesthesia, Dreamzz IVF Center and Women’s Care Hospital, Ahmedabad 382350, Gujarat, India;
| | - Kavita Singh
- Centre for Translational Research, Jiwaji University, Gwalior 474011, Madhya Pradesh, India;
| | - Vimal Patel
- Department of Pharmaceutics, Nirma University, Ahmedabad 382481, Gujarat, India;
| | - Meerambika Mishra
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32611, USA
- Correspondence: (M.M.); (A.K.M.)
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Korea
- Correspondence: (M.M.); (A.K.M.)
| |
Collapse
|
32
|
Spatial Learning Is Associated with Antagonist Outcomes for DNA Methylation and DNA Hydroxymethylation in the Transcriptional Regulation of the Ryanodine Receptor 3. Neural Plast 2021; 2021:9930962. [PMID: 34434232 PMCID: PMC8380497 DOI: 10.1155/2021/9930962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/29/2021] [Accepted: 07/24/2021] [Indexed: 11/18/2022] Open
Abstract
Increasing attention has been drawn to the role that intracellular calcium stores play in neuronal function. Ryr3 is an intracellular calcium channel that contributes to hippocampal long-term potentiation, dendritic spine function, and higher cognitive processes. Interestingly, stimuli that increase neuronal activity upregulate the transcriptional activity of Ryr3 and augment DNA methylation in its proximal promoter. However, if these observations are valid for complex behavioral tasks such as learning and memory remains being evaluated. Relative expression analysis revealed that spatial learning increased the hippocampal levels of Ryr3, whereas mice trained using a visible platform that resulted in no spatial association showed reduced expression. Interestingly, we also observed that specific DNA modifications accompanied these opposite transcriptional changes. Increased DNA methylation was observed in hippocampal samples from spatially trained mice, and increased DNA hydroxymethylation was found in samples from mice trained using a visible platform. Both DNA modifications were not altered in control regions, suggesting that these changes are not generalized, but rather specific modifications associated with this calcium channel's transcriptional regulation. Our two experimental groups underwent the same physical task differing only in the spatial learning component, highlighting the tight relationship between DNA modifications and transcriptional activity in a relevant context such as behavioral training. Our results complement previous observations and suggest that DNA modifications are a reliable signal for the transcriptional activity of Ryr3 and can be useful to understand how conditions such as aging and neuropathological diseases determine altered Ryr3 expression.
Collapse
|
33
|
Singh R, Kaur N, Dhingra N, Kaur T. Protein misfolding, ER Stress and Chaperones: An approach to develop chaperone-based therapeutics for Alzheimer's Disease. Int J Neurosci 2021:1-21. [PMID: 34402740 DOI: 10.1080/00207454.2021.1968859] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a heterogeneous neurodegenerative disorder with complex etiology that eventually leads to dementia. The main culprit of AD is the extracellular deposition of β-amyloid (Aβ) and intracellular neurofibrillary tangles. The protein conformational change and protein misfolding are the key events of AD pathophysiology, therefore endoplasmic reticulum (ER) stress is an apparent consequence. ER, stress-induced unfolded protein response (UPR) mediators (viz. PERK, IRE1, and ATF6) have been reported widely in the AD brain. Considering these factors, preventing proteins misfolding or aggregation of tau or amyloidogenic proteins appears to be the best approach to halt its pathogenesis. Therefore, therapies through chemical and pharmacological chaperones came to light as an alternative for the treatment of AD. Diverse studies have demonstrated 4-phenylbutyric acid (4-PBA) as a potential therapeutic agent in AD. The current review outlined the mechanism of protein misfolding, different etiological features behind the progression of AD, the significance of ER stress in AD, and the potential therapeutic role of different chaperones to counter AD. The study also highlights the gaps in current knowledge of the chaperones-based therapeutic approach and the possibility of developing chaperones as a potential therapeutic agent for AD treatment.
Collapse
Affiliation(s)
- Rimaljot Singh
- Department of Biophysics, Panjab University Chandigarh, India
| | - Navpreet Kaur
- Department of Biophysics, Panjab University Chandigarh, India
| | - Neelima Dhingra
- University Institute of Pharmaceutical Sciences, Panjab University Chandigarh, India
| | - Tanzeer Kaur
- Department of Biophysics, Panjab University Chandigarh, India
| |
Collapse
|
34
|
Liu Y, Yao J, Song Z, Guo W, Sun B, Wei J, Estillore JP, Back TG, Chen SRW. Limiting RyR2 open time prevents Alzheimer's disease-related deficits in the 3xTG-AD mouse model. J Neurosci Res 2021; 99:2906-2921. [PMID: 34352124 DOI: 10.1002/jnr.24936] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/11/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022]
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) progression is driven by a vicious cycle of soluble β-amyloid (Aβ)-induced neuronal hyperactivity. Thus, breaking this vicious cycle by suppressing neuronal hyperactivity may represent a logical approach to stopping AD progression. In support of this, we have recently shown that genetically and pharmacologically limiting ryanodine receptor 2 (RyR2) open time prevented neuronal hyperactivity, memory impairment, dendritic spine loss, and neuronal cell death in a rapid, early onset AD mouse model (5xFAD). Here, we assessed the impact of limiting RyR2 open time on AD-related deficits in a relatively late occurring, slow developing AD mouse model (3xTG-AD) that bears more resemblance (compared to 5xFAD) to that of human AD. Using behavioral tests, long-term potentiation recordings, and Golgi and Nissl staining, we found that the RyR2-E4872Q mutation, which markedly shortens the open duration of the RyR2 channel, prevented learning and memory impairment, defective long-term potentiation, dendritic spine loss, and neuronal cell death in the 3xTG-AD mice. Furthermore, pharmacologically shortening the RyR2 open time with R-carvedilol rescued these AD-related deficits in 3xTG mice. Therefore, limiting RyR2 open time may offer a promising, neuronal hyperactivity-targeted anti-AD strategy.
Collapse
Affiliation(s)
- Yajing Liu
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinjing Yao
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Zhenpeng Song
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Wenting Guo
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Bo Sun
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jinhong Wei
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - John Paul Estillore
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Thomas G Back
- Department of Chemistry, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
35
|
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: 0.8] [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
|
36
|
Mitostasis, Calcium and Free Radicals in Health, Aging and Neurodegeneration. Biomolecules 2021; 11:biom11071012. [PMID: 34356637 PMCID: PMC8301949 DOI: 10.3390/biom11071012] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
Mitochondria play key roles in ATP supply, calcium homeostasis, redox balance control and apoptosis, which in neurons are fundamental for neurotransmission and to allow synaptic plasticity. Their functional integrity is maintained by mitostasis, a process that involves mitochondrial transport, anchoring, fusion and fission processes regulated by different signaling pathways but mainly by the peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α). PGC-1α also favors Ca2+ homeostasis, reduces oxidative stress, modulates inflammatory processes and mobilizes mitochondria to where they are needed. To achieve their functions, mitochondria are tightly connected to the endoplasmic reticulum (ER) through specialized structures of the ER termed mitochondria-associated membranes (MAMs), which facilitate the communication between these two organelles mainly to aim Ca2+ buffering. Alterations in mitochondrial activity enhance reactive oxygen species (ROS) production, disturbing the physiological metabolism and causing cell damage. Furthermore, cytosolic Ca2+ overload results in an increase in mitochondrial Ca2+, resulting in mitochondrial dysfunction and the induction of mitochondrial permeability transition pore (mPTP) opening, leading to mitochondrial swelling and cell death through apoptosis as demonstrated in several neuropathologies. In summary, mitochondrial homeostasis is critical to maintain neuronal function; in fact, their regulation aims to improve neuronal viability and to protect against aging and neurodegenerative diseases.
Collapse
|
37
|
Jurcau A, Simion A. Oxidative Stress in the Pathogenesis of Alzheimer's Disease and Cerebrovascular Disease with Therapeutic Implications. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 19:94-108. [PMID: 32124703 DOI: 10.2174/1871527319666200303121016] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 01/18/2020] [Accepted: 01/28/2020] [Indexed: 12/15/2022]
Abstract
The significant gain in life expectancy led to an increase in the incidence and prevalence of dementia. Although vascular risk factors have long and repeatedly been shown to increase the risk of Alzheimer's Disease (AD), translating these findings into effective preventive measures has failed. In addition, the finding that incident ischemic stroke approximately doubles the risk of a patient to develop AD has been recently reinforced. Current knowledge and pathogenetic hypotheses of AD are discussed. The implication of oxidative stress in the development of AD is reviewed, with special emphasis on its sudden burst in the setting of acute ischemic stroke and the possible link between this increase in oxidative stress and consequent cognitive impairment. Current knowledge and future directions in the prevention and treatment of AD are discussed outlining the hypothesis of a possible beneficial effect of antioxidant treatment in acute ischemic stroke in delaying the onset/progression of dementia.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Faculty of Medicine and Pharmacy, University of Oradea, 410154 Oradea, Romania.,Clinical Municipal Hospital "Dr. G Curteanu", Neurology Ward, Oradea, Romania
| | - Aurel Simion
- Faculty of Medicine and Pharmacy, University of Oradea, 410154 Oradea, Romania.,Clinical Municipal Hospital "Dr. G Curteanu", Neurological Rehabilitation Ward, Oradea, Romania
| |
Collapse
|
38
|
Lalonde C, Grandbois J, Khurana S, Murray A, Tharmalingam S, Tai TC. Late gestational exposure to dexamethasone and fetal programming of abnormal behavior in Wistar Kyoto rats. Brain Behav 2021; 11:e02049. [PMID: 33528889 PMCID: PMC8035474 DOI: 10.1002/brb3.2049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/16/2020] [Accepted: 01/08/2021] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Fetal programming was characterized a few decades ago, explaining the correlation of physiological phenotypes of offspring exposed to early-life stress. High acute or chronic prenatal stress can overwhelm the enzymatic placental barrier, inducing transcriptional changes in the fetus that can result in different adverse behavioral and physiological phenotypes. The current study investigates the impact of exposure to the synthetic glucocorticoid, dexamethasone, during late gestation on behavioral outcomes. METHODS Pregnant Wistar Kyoto rats were given daily subcutaneous injections from gestational days 15-21 of either dexamethasone (0.9% NaCl, 4% EtOH, 100 µg kg-1 day-1 ) or were physically manipulated as naïve controls. Pups were raised normally until 17 weeks of age and underwent the Porsolt swim task and elevated plus maze for depressive and anxiety-like behaviors, respectively. Neural tissue was preserved for genetic analysis using quantitative real-time polymerase chain reaction. RESULTS Statistical analyses show significant disruption of behavior and genetic profiles of offspring exposed to dexamethasone in-utero. Exposed animals spent more time immobile on the swim task and entered open arms of the elevated plus maze more often than their naïve counterparts. In the prefrontal cortex, there was a sex by treatment interaction on gene expression relevant to neural transmission in ryanodine receptor 2, as well as increased gene expression in SNAP25, COMT, and LSAMP in males prenatally exposed to dexamethasone compared with controls. Both dysregulated genes and behavior are linked to decreased anxiety and fear inhibition. CONCLUSION Our results indicate adult offspring exposed to dexamethasone in-utero have a tendency toward passive stress-coping strategies and an inhibition of anxiety on behavioral tasks. Methyltransferase activity, synaptic activity, and cellular processes were disrupted in the prefrontal cortices of these animals. Specifically, genes involved in emotional response pathways were overexpressed, supporting the link between the behavioral and genetic profiles. Combined, we determine that dexamethasone offspring have adaptive predispositions when faced with novel situations, with increased immobility in the swim task and increased exploration on the elevated plus maze.
Collapse
Affiliation(s)
- Christine Lalonde
- Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada.,Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| | - Julie Grandbois
- Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.,Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Sandhya Khurana
- Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada
| | - Alyssa Murray
- Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.,Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Sujeenthar Tharmalingam
- Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada.,Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.,Department of Biology, Laurentian University, Sudbury, ON, Canada.,Department of Chem/Biochem, Laurentian University, Sudbury, ON, Canada
| | - T C Tai
- Biomolecular Sciences, Laurentian University, Sudbury, ON, Canada.,Division of Medical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.,Department of Biology, Laurentian University, Sudbury, ON, Canada.,Department of Chem/Biochem, Laurentian University, Sudbury, ON, Canada
| |
Collapse
|
39
|
Crosstalk among Calcium ATPases: PMCA, SERCA and SPCA in Mental Diseases. Int J Mol Sci 2021; 22:ijms22062785. [PMID: 33801794 PMCID: PMC8000800 DOI: 10.3390/ijms22062785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/20/2022] Open
Abstract
Calcium in mammalian neurons is essential for developmental processes, neurotransmitter release, apoptosis, and signal transduction. Incorrectly processed Ca2+ signal is well-known to trigger a cascade of events leading to altered response to variety of stimuli and persistent accumulation of pathological changes at the molecular level. To counterbalance potentially detrimental consequences of Ca2+, neurons are equipped with sophisticated mechanisms that function to keep its concentration in a tightly regulated range. Calcium pumps belonging to the P-type family of ATPases: plasma membrane Ca2+-ATPase (PMCA), sarco/endoplasmic Ca2+-ATPase (SERCA) and secretory pathway Ca2+-ATPase (SPCA) are considered efficient line of defense against abnormal Ca2+ rises. However, their role is not limited only to Ca2+ transport, as they present tissue-specific functionality and unique sensitive to the regulation by the main calcium signal decoding protein—calmodulin (CaM). Based on the available literature, in this review we analyze the contribution of these three types of Ca2+-ATPases to neuropathology, with a special emphasis on mental diseases.
Collapse
|
40
|
Hori A, Ai T, Isshiki M, Motoi Y, Yano K, Tabe Y, Hattori N, Miida T. Novel Variants in the CLCN1, RYR2, and DCTN1 Found in Elderly Japanese Dementia Patients: A Case Series. Geriatrics (Basel) 2021; 6:geriatrics6010014. [PMID: 33562224 PMCID: PMC7931039 DOI: 10.3390/geriatrics6010014] [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] [Received: 12/06/2020] [Revised: 01/18/2021] [Accepted: 02/04/2021] [Indexed: 11/16/2022] Open
Abstract
Dementia has an enormous impact on medical and financial resources in aging societies like Japan. Diagnosis of dementia can be made by physical and mental examinations, imaging tests, and findings of high abnormal proteins in cerebrospinal fluids. In addition, genetic tests can be performed in neurodegenerative diseases such as Alzheimer’s disease (AD), frontotemporal dementia (FTD), and Parkinson’s disease (PD). In this case series, we presented three cases of dementia with unknown causes who carry novel variants in the genes associated with neurodegenerative diseases. Three patients (Patients 1, 2, and 6) were found by screening 18 dementia patients using a gene panel including 63 genes. The age of onset for Patient 1 was 74 years old, and his father had PD and mother had AD. The age of onset for Patient 2 was 75 years old, and her mother had AD. The age of onset for Patient 6 was 83 years old, and her father, two sisters, and daughter had dementia. The Mini-Mental State Examination produced results of 20, 15, and 22, respectively. The suspected diagnosis by neurological examinations and imaging studies for Patients 1 and 2 was AD, and for Patient 6 was FTD. Patient 1 was treated with donepezil; Patient 2 was treated with donepezil and memantine; and Patient 6 was treated with donepezil, galantamine, and rivastigmine. The three rare variants identified were: CLCN1, encoding a chloride channel, c.2848G>A:p.Glu950Lys (Patient 1); RYR2, encoding a calcium releasing ryanodine receptor, c.13175A>G:p.Lys4392Arg (Patient 2); and DCTN1, encoding a subunit of dynactin, c. 3209G>A:p.Arg1070Gln (Patient 6). The detected variants were interpreted according to the American College of Medical Genetics (ACMG) guidelines. The minor allele frequency for each variant was 0.025%, 0.023%, and 0.0004% in East Asians, respectively. The DCTN1 variant found in Patient 6 might be associated with FTD. Although none of them were previously reported in dementia patients, all variants were classified as variants of unknown significance (VUS). Our report suggests that results of genetic tests in elderly patients with dementia need to be carefully interpreted. Further data accumulation of genotype–phenotype relationships and development of appropriate functional models are warranted.
Collapse
Affiliation(s)
- Atsushi Hori
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8424, Japan; (A.H.); (K.Y.)
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8424, Japan; (M.I.); (Y.T.); (T.M.)
| | - Tomohiko Ai
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8424, Japan; (M.I.); (Y.T.); (T.M.)
- Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: or
| | - Miwa Isshiki
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8424, Japan; (M.I.); (Y.T.); (T.M.)
| | - Yumiko Motoi
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8424, Japan; (Y.M.); (N.H.)
| | - Kouji Yano
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8424, Japan; (A.H.); (K.Y.)
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8424, Japan; (M.I.); (Y.T.); (T.M.)
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo 113-8424, Japan; (Y.M.); (N.H.)
| | - Takashi Miida
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8424, Japan; (M.I.); (Y.T.); (T.M.)
| |
Collapse
|
41
|
Identification of new BACE1 inhibitors for treating Alzheimer's disease. J Mol Model 2021; 27:58. [PMID: 33517514 DOI: 10.1007/s00894-021-04679-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 01/14/2021] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a type of brain disorder, wherein a person experiences gradual memory loss, state of confusion, hallucination, agitation, and personality change. AD is marked by the presence of extracellular amyloid plaques and intracellular neurofibrillary tangles (NFTs) and synaptic losses. Increased cases of AD in recent times created a dire need to discover or identify chemical compounds that can cease the development of AD. This study focuses on finding potential drug molecule(s) active against β-secretase, also known as β-site amyloid precursor protein cleaving enzyme 1 (BACE1). Clustering analysis followed by phylogenetic studies on microarray datasets retrieved from GEO browser showed that BACE1 gene has genetic relatedness with the RCAN1 gene. A ligand library comprising 60 natural compounds retrieved from literature and 25 synthetic compounds collected from DrugBank were screened. Further, 350 analogues of potential parent compounds were added to the library for the docking purposes. Molecular docking studies identified 11-oxotigogenin as the best ligand molecule. The compound showed the binding affinity of - 11.1 Kcal/mole and forms three hydrogen bonds with Trp124, Ile174, and Arg176. The protein-ligand complex was subjected to 25 ns molecular dynamics simulation and the potential energy of the complex was found to be - 1.24579e+06 Kcal/mole. In this study, 11-oxotigogenin has shown promising results against BACE1, which is a leading cause of AD, hence warrants for in vitro and in vivo validation of the same. In addition, in silico identification of 11-oxotigogenin as a potential anti-AD compound paves the way for designing of chemical scaffolds to discover more potent BACE1 inhibitors.Graphical abstract.
Collapse
|
42
|
Quantifying the dose-dependent impact of intracellular amyloid beta in a mathematical model of calcium regulation in xenopus oocyte. PLoS One 2021; 16:e0246116. [PMID: 33508037 PMCID: PMC7842920 DOI: 10.1371/journal.pone.0246116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 01/13/2021] [Indexed: 12/04/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating illness affecting over 40 million people worldwide. Intraneuronal rise of amyloid beta in its oligomeric forms (iAβOs), has been linked to the pathogenesis of AD by disrupting cytosolic Ca2+ homeostasis. However, the specific mechanisms of action are still under debate and intense effort is ongoing to improve our understanding of the crucial steps involved in the mechanisms of AβOs toxicity. We report the development of a mathematical model describing a proposed mechanism by which stimulation of Phospholipase C (PLC) by iAβO, triggers production of IP3 with consequent abnormal release of Ca2+ from the endoplasmic reticulum (ER) through activation of IP3 receptor (IP3R) Ca2+ channels. After validating the model using experimental data, we quantify the effects of intracellular rise in iAβOs on model solutions. Our model validates a dose-dependent influence of iAβOs on IP3-mediated Ca2+ signaling. We investigate Ca2+ signaling patterns for small and large iAβOs doses and study the role of various parameters on Ca2+ signals. Uncertainty quantification and partial rank correlation coefficients are used to better understand how the model behaves under various parameter regimes. Our model predicts that iAβO alter IP3R sensitivity to IP3 for large doses. Our analysis also shows that the upstream production of IP3 can influence Aβ-driven solution patterns in a dose-dependent manner. Model results illustrate and confirm the detrimental impact of iAβOs on IP3 signaling.
Collapse
|
43
|
Chami M. Calcium Signalling in Alzheimer's Disease: From Pathophysiological Regulation to Therapeutic Approaches. Cells 2021; 10:cells10010140. [PMID: 33445705 PMCID: PMC7828109 DOI: 10.3390/cells10010140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
- Mounia Chami
- Laboratory of Excellence DistALZ, INSERM, CNRS, Institute of Molecular and Cellular Pharmacology, Université Côte d'Azur, Sophia-Antipolis, 06560 Valbonne, France
| |
Collapse
|
44
|
Cook DC, Goldstein PA. Non-canonical Molecular Targets for Novel Analgesics: Intracellular Calcium and HCN Channels. Curr Neuropharmacol 2021; 19:1937-1951. [PMID: 33463473 PMCID: PMC9185781 DOI: 10.2174/1570159x19666210119153047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/17/2021] [Indexed: 11/22/2022] Open
Abstract
Pain is a prevalent biopsychosocial condition that poses a significant challenge to healthcare providers, contributes substantially to a disability, and is a major economic burden worldwide. An overreliance on opioid analgesics, which primarily target the μ-opioid receptor, has caused devastating morbidity and mortality in the form of misuse and overdose-related death. Thus, novel analgesic medications are needed that can effectively treat pain and provide an alternative to opioids. A variety of cellular ion channels contribute to nociception, the response of the sensory nervous system to a noxious stimulus that commonly leads to pain. Ion channels involved in nociception may provide a suitable target for pharmacologic modulation to achieve pain relief. This narrative review summarizes the evidence for two ion channels that merit consideration as targets for non-opioid pain medications: ryanodine receptors (RyRs), which are intracellular calcium channels, and hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, which belong to the superfamily of voltage-gated K+ channels. The role of these channels in nociception and neuropathic pain is discussed and suitability as targets for novel analgesics and antihyperalgesics is considered.
Collapse
Affiliation(s)
- Daniel C. Cook
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Peter A. Goldstein
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY 10065, USA
- Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
45
|
Calabrò M, Rinaldi C, Santoro G, Crisafulli C. The biological pathways of Alzheimer disease: a review. AIMS Neurosci 2020; 8:86-132. [PMID: 33490374 PMCID: PMC7815481 DOI: 10.3934/neuroscience.2021005] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer disease is a progressive neurodegenerative disorder, mainly affecting older people, which severely impairs patients' quality of life. In the recent years, the number of affected individuals has seen a rapid increase. It is estimated that up to 107 million subjects will be affected by 2050 worldwide. Research in this area has revealed a lot about the biological and environmental underpinnings of Alzheimer, especially its correlation with β-Amyloid and Tau related mechanics; however, the precise molecular events and biological pathways behind the disease are yet to be discovered. In this review, we focus our attention on the biological mechanics that may lie behind Alzheimer development. In particular, we briefly describe the genetic elements and discuss about specific biological processes potentially associated with the disease.
Collapse
Affiliation(s)
| | | | | | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| |
Collapse
|
46
|
Molecular Dysfunctions of Mitochondria-Associated Membranes (MAMs) in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21249521. [PMID: 33327665 PMCID: PMC7765134 DOI: 10.3390/ijms21249521] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial neurodegenerative pathology characterized by a progressive decline of cognitive functions. Alteration of various signaling cascades affecting distinct subcellular compartment functions and their communication likely contribute to AD progression. Among others, the alteration of the physical association between the endoplasmic reticulum (ER) and mitochondria, also referred as mitochondria-associated membranes (MAMs), impacts various cellular housekeeping functions such as phospholipids-, glucose-, cholesterol-, and fatty-acid-metabolism, as well as calcium signaling, which are all altered in AD. Our review describes the physical and functional proteome crosstalk between the ER and mitochondria and highlights the contribution of distinct molecular components of MAMs to mitochondrial and ER dysfunctions in AD progression. We also discuss potential strategies targeting MAMs to improve mitochondria and ER functions in AD.
Collapse
|
47
|
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: 6.8] [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
|
48
|
Neuronal Autophagy Regulates Presynaptic Neurotransmission by Controlling the Axonal Endoplasmic Reticulum. Neuron 2020; 109:299-313.e9. [PMID: 33157003 PMCID: PMC7837115 DOI: 10.1016/j.neuron.2020.10.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/22/2020] [Accepted: 10/05/2020] [Indexed: 12/21/2022]
Abstract
Neurons are known to rely on autophagy for removal of defective proteins or organelles to maintain synaptic neurotransmission and counteract neurodegeneration. In spite of its importance for neuronal health, the physiological substrates of neuronal autophagy in the absence of proteotoxic challenge have remained largely elusive. We use knockout mice conditionally lacking the essential autophagy protein ATG5 and quantitative proteomics to demonstrate that loss of neuronal autophagy causes selective accumulation of tubular endoplasmic reticulum (ER) in axons, resulting in increased excitatory neurotransmission and compromised postnatal viability in vivo. The gain in excitatory neurotransmission is shown to be a consequence of elevated calcium release from ER stores via ryanodine receptors accumulated in axons and at presynaptic sites. We propose a model where neuronal autophagy controls axonal ER calcium stores to regulate neurotransmission in healthy neurons and in the brain. Neuronal autophagy controls the endoplasmic reticulum (ER) in axons Loss of neuronal autophagy leads to increased excitatory neurotransmission Increased neurotransmission is due to elevated calcium release from ER stores
Collapse
|
49
|
|
50
|
Pizzo P, Basso E, Filadi R, Greotti E, Leparulo A, Pendin D, Redolfi N, Rossini M, Vajente N, Pozzan T, Fasolato C. Presenilin-2 and Calcium Handling: Molecules, Organelles, Cells and Brain Networks. Cells 2020; 9:E2166. [PMID: 32992716 PMCID: PMC7601421 DOI: 10.3390/cells9102166] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Presenilin-2 (PS2) is one of the three proteins that are dominantly mutated in familial Alzheimer's disease (FAD). It forms the catalytic core of the γ-secretase complex-a function shared with its homolog presenilin-1 (PS1)-the enzyme ultimately responsible of amyloid-β (Aβ) formation. Besides its enzymatic activity, PS2 is a multifunctional protein, being specifically involved, independently of γ-secretase activity, in the modulation of several cellular processes, such as Ca2+ signalling, mitochondrial function, inter-organelle communication, and autophagy. As for the former, evidence has accumulated that supports the involvement of PS2 at different levels, ranging from organelle Ca2+ handling to Ca2+ entry through plasma membrane channels. Thus FAD-linked PS2 mutations impact on multiple aspects of cell and tissue physiology, including bioenergetics and brain network excitability. In this contribution, we summarize the main findings on PS2, primarily as a modulator of Ca2+ homeostasis, with particular emphasis on the role of its mutations in the pathogenesis of FAD. Identification of cell pathways and molecules that are specifically targeted by PS2 mutants, as well as of common targets shared with PS1 mutants, will be fundamental to disentangle the complexity of memory loss and brain degeneration that occurs in Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Paola Pizzo
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Emy Basso
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Alessandro Leparulo
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
| | - Diana Pendin
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Nelly Redolfi
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
| | - Michela Rossini
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
| | - Nicola Vajente
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
- Neuroscience Institute, Italian National Research Council (CNR), Via U. Bassi 58/B, 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), Via G. Orus 2B, 35131 Padua, Italy
| | - Cristina Fasolato
- Department of Biomedical Sciences, University of Padua, Via U. Bassi 58/B, 35131 Padua, Italy; (E.B.); (R.F.); (E.G.); (A.L.); (D.P.); (N.R.); (M.R.); (N.V.); (T.P.)
| |
Collapse
|