Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(20)2023 Oct 21.
Article in English | MEDLINE | ID: mdl-37895105

ABSTRACT

The transient receptor potential cation channel, subfamily C, member 6 (TRPC6), has been believed to adjust the formation of an excitatory synapse. The positive regulation of TRPC6 engenders synapse enlargement and improved learning and memory in animal models. TRPC6 is involved in different synaptoprotective signaling pathways, including antagonism of N-methyl-D-aspartate receptor (NMDAR), activation of brain-derived neurotrophic factor (BDNF) and postsynaptic store-operated calcium entry. Positive regulation of TRPC6 channels has been repeatedly shown to be good for memory formation and storage. TRPC6 is mainly expressed in the hippocampus, particularly in the dentate granule cells, cornu Ammonis 3 (CA3) pyramidal cells and gamma-aminobutyric acid (GABA)ergic interneurons. It has been observed that TRPC6 agonists have a great influence on animal behavior including memory formation and storage The purpose of this review is to collect the available information on the role of TRPC6 in memory formation in various parts of the brain to understand how TRPC6-specific pharmaceutical agents will affect memory in distinct parts of the central nervous system (CNS).


Subject(s)
Neurons , TRPC Cation Channels , Animals , TRPC6 Cation Channel , TRPC Cation Channels/metabolism , Neurons/metabolism , Synapses/metabolism , Behavior, Animal , Calcium/metabolism
2.
Curr Issues Mol Biol ; 44(11): 5191-5208, 2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36354665

ABSTRACT

Alzheimer's disease (AD) is characterized by synaptic dysfunction, which is expressed through the loss of dendritic spines and changes in their morphology. Pharmacological compounds that are able to protect spines in the AD brain are suggested to be novel drugs that would be able to slow down the disease progression. We have recently shown that a positive modulator of transient receptor potential cation channel subfamily C member 6 (TRPC6), the compound N-(2-chlorophenyl)-2-(4-phenylpiperazine-1-yl) acetamide (51164), causes the upregulation of postsynaptic neuronal store-operated calcium entry, maintains mushroom spine percentage, and recovers synaptic plasticity in amyloidogenic mouse models of Alzheimer's disease. Here, using confocal microscopy and calcium imaging methods, we present the experimental data indicating that 51164 possesses an alternative mechanism of action. We demonstrated that 51164 can increase the mushroom spine percentage in neurons with the downregulated activity of TRPC6-dependent neuronal store-operated calcium entry. Moreover, we report the binding of 51164 to G-actin in silico. We observed that 51164 interacts with Lys 336, Asp157, and Ser14 of G-actin, amino acids involved in the stabilization/polymerization of the G-actin structure. We showed that interactions of 51164 with G-actin are much stronger in comparison to the well-characterized F-actin stabilizing and polymerizing drug, jasplakinolide. The obtained results suggest an alternative protective mechanism of 51164 that is related to the preservation of actin filaments in vitro.

3.
Int J Mol Sci ; 23(21)2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36362339

ABSTRACT

Synapse loss in the brain of Alzheimer's disease patients correlates with cognitive dysfunctions. Drugs that limit synaptic loss are promising pharmacological agents. The transient receptor potential cation channel, subfamily C, member 6 (TRPC6) regulates the formation of an excitatory synapse. Positive regulation of TRPC6 results in increased synapse formation and enhances learning and memory in animal models. The novel selective TRPC6 agonist, 3-(3-,4-Dihydro-6,7-dimethoxy-3,3-dimethyl-1-isoquinolinyl)-2H-1-benzopyran-2-one, has recently been identified. Here we present in silico, in vitro, ex vivo, pharmacokinetic and in vivo studies of this compound. We demonstrate that it binds to the extracellular agonist binding site of the human TRPC6, protects hippocampal mushroom spines from amyloid toxicity in vitro, efficiently recovers synaptic plasticity in 5xFAD brain slices, penetrates the blood-brain barrier and recovers cognitive deficits in 5xFAD mice. We suggest that C20 might be recognized as the novel TRPC6-selective drug suitable to treat synaptic deficiency in Alzheimer's disease-affected hippocampal neurons.


Subject(s)
Alzheimer Disease , Mice , Animals , Humans , TRPC6 Cation Channel/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Blood-Brain Barrier/metabolism , Memory Disorders/drug therapy , Memory Disorders/metabolism , Hippocampus/metabolism , Mice, Transgenic , Disease Models, Animal , Amyloid beta-Peptides/metabolism
4.
IBRO Neurosci Rep ; 12: 90-97, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35079728

ABSTRACT

Calcium/calmodulin-dependent protein kinase II (CaMKII) and neuronal store-operated calcium entry (nSOCE) have been implicated in the development of Alzheimer's disease (AD). nSOCE is involved in regulation of dendritic spine shape, particularly in stability of mushroom spines that play role in formation of strong synapses. CaMKII is involved in regulation of induction of long-term potentiation, that is needed for shaping of memory. In the present study, we demonstrated that inhibition of kinase activity of CaMKII by KN-62 decreases nSOCE amplitude in soma of primary hippocampal neurons. We have shown that knockdown of CaMKIIß leads to the downregulation of nSOCE in dendritic spines. In agreement with previously published data, we have also observed that CaMKIIß knockdown causes mushroom spine loss in primary hippocampal culture. The effect of CaMKIIß knockdown on the nSOCE may be associated with a decrease of dendritic spine head size.

5.
Int J Mol Sci ; 22(24)2021 Dec 19.
Article in English | MEDLINE | ID: mdl-34948414

ABSTRACT

Store-operated calcium entry (SOCE) constitutes a fine-tuning mechanism responsible for the replenishment of intracellular stores. Hippocampal SOCE is regulated by store-operated channels (SOC) organized in tripartite complex TRPC6/ORAI2/STIM2. It is suggested that in neurons, SOCE maintains intracellular homeostatic Ca2+ concentration at resting conditions and is needed to support the structure of dendritic spines. Recent evidence suggests that positive modulators of SOC are prospective drug candidates to treat Alzheimer's disease (AD) at early stages. Although STIM2 and ORAI2 are definitely involved in the regulation of nSOC amplitude and a play major role in AD pathogenesis, growing evidence suggest that it is not easy to target these proteins pharmacologically. Existing positive modulators of TRPC6 are unsuitable for drug development due to either bad pharmacokinetics or side effects. Thus, we concentrate the review on perspectives to develop specific nSOC modulators based on available 3D structures of TRPC6, ORAI2, and STIM2. We shortly describe the structural features of existing models and the methods used to prepare them. We provide commonly used steps applied for drug design based on 3D structures of target proteins that might be used to develop novel AD preventing therapy.


Subject(s)
Alzheimer Disease/drug therapy , Drug Design , ORAI2 Protein/metabolism , Stromal Interaction Molecule 2/metabolism , TRPC6 Cation Channel/metabolism , Alzheimer Disease/metabolism , Animals , Drug Discovery , Humans , ORAI2 Protein/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Stromal Interaction Molecule 2/chemistry , Synapses/drug effects , Synapses/metabolism , TRPC6 Cation Channel/chemistry
6.
Cells ; 9(11)2020 10 24.
Article in English | MEDLINE | ID: mdl-33114455

ABSTRACT

Alzheimer's disease and cerebral ischemia are among the many causative neurodegenerative diseases that lead to disabilities in the middle-aged and elderly population. There are no effective disease-preventing therapies for these pathologies. Recent in vitro and in vivo studies have revealed the TRPC6 channel to be a promising molecular target for the development of neuroprotective agents. TRPC6 channel is a non-selective cation plasma membrane channel that is permeable to Ca2+. Its Ca2+-dependent pharmacological effect is associated with the stabilization and protection of excitatory synapses. Downregulation as well as upregulation of TRPC6 channel functions have been observed in Alzheimer's disease and brain ischemia models. Thus, in order to protect neurons from Alzheimer's disease and cerebral ischemia, proper TRPC6 channels modulators have to be used. TRPC6 channels modulators are an emerging research field. New chemical structures modulating the activity of TRPC6 channels are being currently discovered. The recent publication of the cryo-EM structure of TRPC6 channels should speed up the discovery process even more. This review summarizes the currently available information about potential drug candidates that may be used as basic structures to develop selective, highly potent TRPC6 channel modulators to treat neurodegenerative disorders, such as Alzheimer's disease and cerebral ischemia.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Brain Ischemia/etiology , Brain Ischemia/metabolism , TRPC6 Cation Channel/deficiency , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Disease Management , Disease Susceptibility , Drug Discovery , Gene Expression Regulation/drug effects , Humans , Molecular Targeted Therapy , Risk Factors , Signal Transduction/drug effects , TRPC6 Cation Channel/antagonists & inhibitors , TRPC6 Cation Channel/genetics , TRPC6 Cation Channel/metabolism
7.
Curr Alzheimer Res ; 17(4): 344-354, 2020.
Article in English | MEDLINE | ID: mdl-32469698

ABSTRACT

Despite decades of research and effort, there is still no effective disease-modifying treatment for Alzheimer's Disease (AD). Most of the recent AD clinical trials were targeting amyloid pathway, but all these trials failed. Although amyloid pathology is a hallmark and defining feature of AD, targeting the amyloid pathway has been very challenging due to low efficacy and serious side effects. Alternative approaches or mechanisms for our understanding of the major cause of memory loss in AD need to be considered as potential therapeutic targets. Increasing studies suggest that Ca2+ dysregulation in AD plays an important role in AD pathology and is associated with other AD abnormalities, such as excessive inflammation, increased ROS, impaired autophagy, neurodegeneration, synapse, and cognitive dysfunction. Ca2+ dysregulation in cytosolic space, Endoplasmic Reticulum (ER) and mitochondria have been reported in the context of various AD models. Drugs or strategies, to correct the Ca2+ dysregulation in AD, have been demonstrated to be promising as an approach for the treatment of AD in preclinical models. This review will discuss the mechanisms of Ca2+ dysregulation in AD and associated pathology and discuss potential approaches or strategies to develop novel drugs for the treatment of AD by targeting Ca2+ dysregulation.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Calcium Channel Blockers/therapeutic use , Calcium/metabolism , Animals , Calcium Channel Blockers/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Humans , Intracellular Calcium-Sensing Proteins/antagonists & inhibitors , Intracellular Calcium-Sensing Proteins/metabolism , Treatment Outcome
8.
Curr Alzheimer Res ; 17(12): 1065-1071, 2020.
Article in English | MEDLINE | ID: mdl-33463468

ABSTRACT

Alzheimer disease is the dominant form of elderly dementia. Today all clinical trials that target ß-amyloid have failed to indicate that ß-amyloid may not be a causative agent in AD pathogenesis. Thus there is a need to search for alternative ways to treat AD patients. Neuronal store-operated calcium entry is a fine-tuning mechanism that regulates intracellular Ca2+ content. Recent evidence suggests that store-operated calcium channels may be targeted with pharmacological agents in order to prevent synapse loss, recover long-term potentiation and change behavior. Current mini-review discusses basic chemical structures that modulate intracellular calcium dysbalance via targeting store-operated calcium channels and their applicability as anti-AD pharmacological agents.


Subject(s)
Alzheimer Disease/pathology , Calcium/metabolism , Hippocampus/metabolism , Pharmacological Phenomena , Synapses/pathology , Animals , Humans , Mice , Neurons/pathology
9.
Neuroscience ; 410: 118-127, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31055008

ABSTRACT

Alzheimer's disease (AD) is the neurodegenerative disorder with no cure. Recent studies suggest that dysregulated postsynaptic store-operated calcium entry (nSOCE) may underlie mushroom spine loss that is related to AD pathology. In the present study we observed that PSEN1ΔE9 familial AD (FAD) mutation causes mushroom spine loss in hippocampal neuronal cultures. We also demonstrated that amplitude of TRPC6-mediated nSOCE is increased in PSEN1ΔE9-expressing neurons and we suggested that inhibition of nSOCE may help to rescue synaptic defects in this model. We further established that nSOCE antagonist EVP4593 decreases PSEN1ΔE9-mediated nSOCE upregulation and rescues mushroom spines in PSEN1ΔE9-expressing neurons. Obtained results further highlight the connection between dysregulation of endoplasmic reticulum calcium signaling and synaptic loss in AD and suggest that calcium signaling modulators may have a therapeutic value for treatment of memory loss in AD.


Subject(s)
Alzheimer Disease/metabolism , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Neurons/metabolism , Phenyl Ethers/pharmacology , Presenilin-1/biosynthesis , Quinazolines/pharmacology , Alzheimer Disease/genetics , Animals , Calcium Channel Blockers/pharmacology , Calcium-Binding Proteins/genetics , Cells, Cultured , Gene Expression , Hippocampus/drug effects , Hippocampus/metabolism , Membrane Proteins/genetics , Mice , Mice, Transgenic , Mutation/physiology , Neurons/drug effects , Presenilin-1/genetics , Treatment Outcome
10.
Mol Pharmacol ; 95(4): 337-348, 2019 04.
Article in English | MEDLINE | ID: mdl-30696719

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder that is the major cause of dementia in the elderly. There is no cure against AD. We have recently discovered a novel transient receptor potential canonical 6 (TRPC6)-mediated intracellular signaling pathway that regulates the stability of dendritic spines and plays a role in memory formation. We have previously shown that TRPC6 agonists exert beneficial effects in models of AD and may serve as lead compounds for development of AD therapeutic agents. In the current study, we used the Clarivate Analytics Integrity database to search for additional TRPC6 agonists. We selected four compounds to study as potential neuroprotective agents. We applied bioinformatics analyses to test the basic pharmacological properties of the selected compounds. We performed in vitro screening of these compounds to validate their ability to protect mushroom spines from amyloid toxicity and determined that two of these compounds exert neuroprotective effects in the nanomolar concentration range. We have chosen one of these compounds [piperazine (PPZ)] for further testing. In agreement with previously published data, we have shown that PPZ potentiates TRPC6 channels. We demonstrated that the neuroprotective mechanism of the investigated PPZ is based on activation of neuronal store-operated calcium entry in spines. We have shown that PPZ restores long-term potentiation induction in 6-month-old 5xFAD mouse hippocampal slices. The obtained results suggest that PPZ and its derivatives are potential lead molecules for development of AD therapeutic agents.


Subject(s)
Alzheimer Disease/drug therapy , Piperazines/pharmacology , Alzheimer Disease/metabolism , Animals , Calcium Signaling/drug effects , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , TRPC Cation Channels/metabolism
11.
Antioxid Redox Signal ; 29(12): 1176-1188, 2018 10 20.
Article in English | MEDLINE | ID: mdl-29890840

ABSTRACT

SIGNIFICANCE: Calcium (Ca2+) hypothesis of Alzheimer's disease (AD) gains popularity. It points to new signaling pathways that may underlie AD pathogenesis. Based on calcium hypothesis, novel targets for the development of potential AD therapies are identified. Recent Advances: Recently, the key role of neuronal store-operated calcium entry (nSOCE) in the development of AD has been described. Correct regulation of nSOCE is necessary for the stability of postsynaptic contacts to preserve the memory formation. Molecular identity of hippocampal nSOCE is defined. Perspective nSOCE-activating molecule, prototype of future anti-AD drugs, is described. CRITICAL ISSUES: Endoplasmic reticulum Ca2+ overload happens in many but not in all AD models. The nSOCE targeting therapy described in this review may not be universally applicable. FUTURE DIRECTIONS: There is a need to determine whether AD is a syndrome with one critical signaling pathway that initiates pathology, or it is a disorder with many different signaling pathways that are disrupted simultaneously or one after each other. It is necessary to validate applicability of nSOCE-activating therapy for the development of anti-AD medication. There is an experimental correlation between downregulated nSOCE and disrupted postsynaptic contacts in AD mouse models. Signaling mechanisms downstream of nSOCE which are responsible for the regulation of stability of postsynaptic contacts have to be discovered. That will bring new targets for the development of AD-preventing therapies. Antioxid. Redox Signal. 29, 1176-1188.


Subject(s)
Alzheimer Disease/metabolism , Calcium Signaling , Calcium/metabolism , Alzheimer Disease/pathology , Animals , Endoplasmic Reticulum/chemistry , Endoplasmic Reticulum/metabolism , Humans
13.
Cell Calcium ; 70: 87-94, 2018 03.
Article in English | MEDLINE | ID: mdl-28728834

ABSTRACT

Calcium (Ca2+) is a ubiquitous second messenger that regulates various activities in eukaryotic cells. Especially important role calcium plays in excitable cells. Neurons require extremely precise spatial-temporal control of calcium-dependent processes because they regulate such vital functions as synaptic plasticity. Recent evidence indicates that neuronal calcium signaling is abnormal in many of neurodegenerative disorders such as Alzheimer's disease (AD), Huntington's disease (HD) and Parkinson's disease (PD). These diseases represent a major medical, social, financial and scientific problem, but despite enormous research efforts, they are still incurable and only symptomatic relief drugs are available. Thus, new approaches and targets are needed. This review highlight neuronal calcium-signaling abnormalities in these diseases, with particular emphasis on the role of neuronal store-operated Ca2+ entry (SOCE) pathway and its potential relevance as a therapeutic target for treatment of neurodegeneration.


Subject(s)
Calcium Signaling , Neurodegenerative Diseases/genetics , Animals , Calcium/metabolism , Humans , Models, Biological
14.
Sci Rep ; 7(1): 17625, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29247211

ABSTRACT

Mushroom spines form strong synaptic contacts and are essential for memory storage. We have previously demonstrated that neuronal store-operated calcium entry (nSOC) in hippocampal neurons is regulated by STIM2 protein. This pathway plays a key role in stability of mushroom spines and is compromised in different mice models of Alzheimer's disease (AD). Actin was thought to be the sole cytoskeleton compartment presented in dendritic spines, however, recent studies demonstrated that dynamic microtubules with EB3 capped plus-ends transiently enter spines. We showed that STIM2 forms an endoplasmic reticulum (ER) Ca2+ -dependent complex with EB3 via Ser-x-Ile-Pro aminoacid motif and that disruption of STIM2-EB3 interaction resulted in loss of mushroom spines in hippocampal neurons. Overexpression of EB3 causes increase of mushroom spines fraction and is able to restore their deficiency in hippocampal neurons obtained from PS1-M146V-KI AD mouse model. STIM2 overexpression failed to restore mushroom dendritic spines after EB3 knockdown, while in contrast EB3 overexpression rescued loss of mushroom spines resulting from STIM2 depletion. We propose that EB3 is involved in regulation of dendritic spines morphology, in part due to its association with STIM2, and that modulation of EB3 expression is a potential way to overcome synaptic loss during AD.


Subject(s)
Alzheimer Disease/pathology , Calcium/metabolism , Dendritic Spines/pathology , Hippocampus/metabolism , Microtubule-Associated Proteins/metabolism , Stromal Interaction Molecule 2/metabolism , Animals , Calcium Signaling/physiology , Cells, Cultured , Disease Models, Animal , Gene Knock-In Techniques , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Synapses/physiology
15.
Biochem Biophys Res Commun ; 483(4): 998-1004, 2017 02 19.
Article in English | MEDLINE | ID: mdl-27641664

ABSTRACT

Alzheimer's disease (AD) is the disease of lost memories. Synaptic loss is a major reason for memory defects in AD. Signaling pathways involved in memory loss in AD are under intense investigation. The role of deranged neuronal calcium (Ca2+) signaling in synaptic loss in AD is described in this review. Familial AD (FAD) mutations in presenilins are linked directly with synaptic Ca2+ signaling abnormalities, most likely by affecting endoplasmic reticulum (ER) Ca2+ leak function of presenilins. Excessive ER Ca2+ release via type 2 ryanodine receptors (RyanR2) is observed in AD spines due to increase in expression and function of RyanR2. Store-operated Ca2+ entry (nSOC) pathway is disrupted in AD spines due to downregulation of STIM2 protein. Because of these Ca2+ signaling abnormalities, a balance in activities of Ca2+-calmodulin-dependent kinase II (CaMKII) and Ca2+-dependent phosphatase calcineurin (CaN) is shifted at the synapse, tilting a balance between long-term potentiation (LTP) and long-term depression (LTD) synaptic mechanisms. As a result, synapses are weakened and eliminated in AD brains by LTD mechanism, causing memory loss. Targeting synaptic calcium signaling pathways offers opportunity for development of AD therapeutic agents.


Subject(s)
Alzheimer Disease/metabolism , Calcium/metabolism , Homeostasis , Neurons/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Animals , Calcineurin/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Humans , Long-Term Potentiation , Long-Term Synaptic Depression , Ryanodine Receptor Calcium Release Channel/metabolism
16.
Neurodegener Dis Manag ; 5(5): 395-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26477700

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder that primarily compromises memory formation and storage. Several hypotheses regarding the pathogenesis of AD have been proposed; however, no cure is available to date. Here we describe the calcium hypothesis of AD, which is gaining popularity. We present data supporting this hypothesis and focus on a recently discovered calcium-signaling pathway that is dysregulated in AD and propose targets for the development of disease-modifying therapies.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Homeostasis/drug effects , Homeostasis/physiology , Animals , Humans
17.
Mol Neurodegener ; 10: 37, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26275606

ABSTRACT

BACKGROUND: Alzheimer disease (AD) is a disease of lost memories. Mushroom postsynaptic spines play a key role in memory storage, and loss of mushroom spines has been proposed to be linked to memory loss in AD. Generation of amyloidogenic peptides and accumulation of amyloid plaques is one of the pathological hallmarks of AD. It is important to evaluate effects of amyloid on stability of mushroom spines. RESULTS: In this study we used in vitro and in vivo models of amyloid synaptotoxicity to investigate effects of amyloid peptides on hippocampal mushroom spines. We discovered that application of Aß42 oligomers to hippocampal cultures or injection of Aß42 oligomers directly into hippocampal region resulted in reduction of mushroom spines and activity of synaptic calcium-calmodulin-dependent kinase II (CaMKII). We further discovered that expression of STIM2 protein rescued CaMKII activity and protected mushroom spines from amyloid toxicity in vitro and in vivo. CONCLUSIONS: Obtained results suggest that downregulation of STIM2-dependent stability of mushroom spines and reduction in activity of synaptic CaMKII is a mechanism of hippocampal synaptic loss in AD model of amyloid synaptotoxicity and that modulators/activators of this pathway may have a potential therapeutic value for treatment of AD.


Subject(s)
Amyloid beta-Peptides/toxicity , CA1 Region, Hippocampal/drug effects , Dendrites/drug effects , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/physiology , Peptide Fragments/toxicity , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Cells, Cultured , Dendrites/ultrastructure , Gene Expression Regulation , Genes, Reporter , Genes, Synthetic , Genetic Vectors/pharmacology , Humans , Injections , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stromal Interaction Molecule 2 , Synapses/enzymology , Transfection
18.
Semin Cell Dev Biol ; 40: 127-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25846864

ABSTRACT

Neurodegenerative disorders, such as spinocerebellar ataxias (SCAs) and Alzheimer's disease (AD) represent a huge scientific and medical question, but the molecular mechanisms of these diseases are still not clear. There is increasing evidence that neuronal calcium signaling is abnormal in many neurodegenerative disorders. Abnormal neuronal calcium release from the endoplasmic reticulum may result in disturbances of cell homeostasis, synaptic dysfunction, and eventual cell death. Neuronal loss is observed in most cases of neurodegenerative diseases. Recent experimental evidence supporting the role of neuronal calcium signaling in the pathogenesis of SCAs and AD is discussed in this review.


Subject(s)
Alzheimer Disease/metabolism , Calcium Signaling , Spinocerebellar Ataxias/metabolism , Alzheimer Disease/pathology , Animals , Humans , Spinocerebellar Ataxias/pathology , Synapses/pathology
19.
J Alzheimers Dis ; 45(2): 561-80, 2015.
Article in English | MEDLINE | ID: mdl-25589721

ABSTRACT

Alzheimer's disease (AD) and aging result in impaired ability to store memories, but the cellular mechanisms responsible for these defects are poorly understood. Presenilin 1 (PS1) mutations are responsible for many early-onset familial AD (FAD) cases. The phenomenon of hippocampal long-term potentiation (LTP) is widely used in studies of memory formation and storage. Recent data revealed long-term LTP maintenance (L-LTP) is impaired in PS1-M146V knock-in (KI) FAD mice. To understand the basis for this phenomenon, in the present study we analyzed structural synaptic plasticity in hippocampal cultures from wild type (WT) and KI mice. We discovered that exposure to picrotoxin induces formation of mushroom spines in both WT and KI cultures, but the maintenance of mushroom spines is impaired in KI neurons. This maintenance defect can be explained by an abnormal firing pattern during the consolidation phase of structural plasticity in KI neurons. Reduced frequency of neuronal firing in KI neurons is caused by enhanced calcium-induced calcium release (CICR), enhanced activity of calcium-activated potassium channels, and increased afterhyperpolarization. As a result, "consolidation" pattern of neuronal activity converted to "depotentiation" pattern of neuronal activity in KI neurons. Consistent with this model, we demonstrated that pharmacological inhibitors of CICR (dantrolene), of calcium-activated potassium channels (apamin), and of calcium-dependent phosphatase calcineurin (FK506) are able to rescue structural plasticity defects in KI neurons. Furthermore, we demonstrate that incubation with dantrolene or apamin also rescued L-LTP defects in KI hippocampal slices, suggesting a role for a similar mechanism. This proposed mechanism may be responsible for memory defects in AD but also for age-related memory decline.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Calcium Signaling/physiology , Hippocampus/pathology , Neuronal Plasticity/physiology , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Animals , Calcium Channel Blockers/pharmacology , Calcium Signaling/genetics , Cells, Cultured , Central Nervous System Stimulants/pharmacology , Dendritic Spines/genetics , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neuronal Plasticity/drug effects , Neurons/pathology , Picrotoxin/pharmacology , Presenilin-1/genetics , Synaptic Potentials/drug effects , Synaptic Potentials/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...