Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 298
Filter
1.
Neurosci Bull ; 38(2): 135-148, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34542799

ABSTRACT

The family of voltage-gated potassium Kv2 channels consists of the Kv2.1 and Kv2.2 subtypes. Kv2.1 is constitutively highly phosphorylated in neurons and its function relies on its phosphorylation state. Whether the function of Kv2.2 is also dependent on its phosphorylation state remains unknown. Here, we investigated whether Kv2.2 channels can be phosphorylated by protein kinase C (PKC) and examined the effects of PKC-induced phosphorylation on their activity and function. Activation of PKC inhibited Kv2.2 currents and altered their steady-state activation in HEK293 cells. Point mutations and specific antibodies against phosphorylated S481 or S488 demonstrated the importance of these residues for the PKC-dependent modulation of Kv2.2. In layer II pyramidal neurons in cortical slices, activation of PKC similarly regulated native Kv2.2 channels and simultaneously reduced the frequency of action potentials. In conclusion, this study provides the first evidence to our knowledge that PKC-induced phosphorylation of the Kv2.2 channel controls the excitability of cortical pyramidal neurons.


Subject(s)
Protein Kinase C , Pyramidal Cells/enzymology , Shab Potassium Channels , Action Potentials , HEK293 Cells , Humans , Protein Kinase C/metabolism , Shab Potassium Channels/genetics
2.
Neuroscience Bulletin ; (6): 135-148, 2022.
Article in English | WPRIM (Western Pacific) | ID: wpr-922667

ABSTRACT

The family of voltage-gated potassium Kv2 channels consists of the Kv2.1 and Kv2.2 subtypes. Kv2.1 is constitutively highly phosphorylated in neurons and its function relies on its phosphorylation state. Whether the function of Kv2.2 is also dependent on its phosphorylation state remains unknown. Here, we investigated whether Kv2.2 channels can be phosphorylated by protein kinase C (PKC) and examined the effects of PKC-induced phosphorylation on their activity and function. Activation of PKC inhibited Kv2.2 currents and altered their steady-state activation in HEK293 cells. Point mutations and specific antibodies against phosphorylated S481 or S488 demonstrated the importance of these residues for the PKC-dependent modulation of Kv2.2. In layer II pyramidal neurons in cortical slices, activation of PKC similarly regulated native Kv2.2 channels and simultaneously reduced the frequency of action potentials. In conclusion, this study provides the first evidence to our knowledge that PKC-induced phosphorylation of the Kv2.2 channel controls the excitability of cortical pyramidal neurons.


Subject(s)
Humans , Action Potentials , HEK293 Cells , Protein Kinase C/metabolism , Pyramidal Cells/enzymology , Shab Potassium Channels/genetics
3.
Mol Brain ; 14(1): 139, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34507588

ABSTRACT

Chronic pain easily leads to concomitant mood disorders, and the excitability of anterior cingulate cortex (ACC) pyramidal neurons (PNs) is involved in chronic pain-related anxiety. However, the mechanism by which PNs regulate pain-related anxiety is still unknown. The GABAergic system plays an important role in modulating neuronal activity. In this paper, we aimed to study how the GABAergic system participates in regulating the excitability of ACC PNs, consequently affecting chronic inflammatory pain-related anxiety. A rat model of CFA-induced chronic inflammatory pain displayed anxiety-like behaviors, increased the excitability of ACC PNs, and reduced inhibitory presynaptic transmission; however, the number of GAD65/67 was not altered. Interestingly, intra-ACC injection of the GABAAR agonist muscimol relieved anxiety-like behaviors but had no effect on chronic inflammatory pain. Intra-ACC injection of the GABAAR antagonist picrotoxin induced anxiety-like behaviors but had no effect on pain in normal rats. Notably, chemogenetic activation of GABAergic neurons in the ACC alleviated chronic inflammatory pain and pain-induced anxiety-like behaviors, enhanced inhibitory presynaptic transmission, and reduced the excitability of ACC PNs. Chemogenetic inhibition of GABAergic neurons in the ACC led to pain-induced anxiety-like behaviors, reduced inhibitory presynaptic transmission, and enhanced the excitability of ACC PNs but had no effect on pain in normal rats. We demonstrate that the GABAergic system mediates a reduction in inhibitory presynaptic transmission in the ACC, which leads to enhanced excitability of pyramidal neurons in the ACC and is associated with chronic inflammatory pain-related anxiety.


Subject(s)
Anxiety/physiopathology , Chronic Pain/physiopathology , GABAergic Neurons/physiology , Gyrus Cinguli/physiopathology , Inflammation/psychology , Pyramidal Cells/physiology , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Anxiety/etiology , Central Nervous System Sensitization/drug effects , Chronic Pain/psychology , Clozapine/therapeutic use , Freund's Adjuvant/toxicity , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Agonists/therapeutic use , GABA-A Receptor Antagonists/administration & dosage , GABA-A Receptor Antagonists/pharmacology , GABA-A Receptor Antagonists/toxicity , GABAergic Neurons/enzymology , Genetic Vectors/pharmacology , Inflammation/chemically induced , Inflammation/physiopathology , Injections , Interneurons/drug effects , Male , Muscimol/administration & dosage , Muscimol/pharmacology , Muscimol/therapeutic use , Open Field Test , Pain Threshold/drug effects , Patch-Clamp Techniques , Picrotoxin/toxicity , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Pyramidal Cells/enzymology , Rats , Rats, Sprague-Dawley
4.
Nutrients ; 12(8)2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32824513

ABSTRACT

Pycnogenol® (an extract of the bark of French maritime pine tree) is used for dietary supplement and known to have excellent antioxidative efficacy. However, there are few reports on neuroprotective effect of Pycnogenol® supplementation and its mechanisms against ischemic injury following transient forebrain ischemia (TFI) in gerbils. Now, we examined neuroprotective effect and its mechanisms of Pycnogenol® in the gerbils with 5-min TFI, which evokes a significant death (loss) of pyramidal cells located in the cornu ammonis (CA1) region of gerbil hippocampus from 4-5 days post-TFI. Gerbils were pretreated with 30, 40, and 50 mg/kg of Pycnogenol® once a day for 7 days before TFI surgery. Treatment with 50 mg/kg, not 30 or 40 mg/kg, of Pycnogenol® potently protected learning and memory, as well as CA1 pyramidal cells, from ischemic injury. Treatment with 50 mg/kg Pycnogenol® significantly enhanced immunoreactivity of antioxidant enzymes (superoxide dismutases and catalase) in the pyramidal cells before and after TFI induction. Furthermore, the treatment significantly reduced the generation of superoxide anion, ribonucleic acid oxidation and lipid peroxidation in the pyramidal cells. Moreover, interestingly, its neuroprotective effect was abolished by administration of sodium azide (a potent inhibitor of SODs and catalase activities). Taken together, current results clearly indicate that Pycnogenol® supplementation can prevent neurons from ischemic stroke through its potent antioxidative role.


Subject(s)
Antioxidants , CA1 Region, Hippocampal/cytology , Dietary Supplements , Flavonoids/administration & dosage , Flavonoids/pharmacology , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/pathology , Memory Disorders/etiology , Memory Disorders/prevention & control , Neuroprotective Agents , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Animals , Catalase/metabolism , Disease Models, Animal , Gerbillinae , Lipid Peroxidation/drug effects , Male , Pyramidal Cells/enzymology , Superoxide Dismutase/metabolism
5.
Mol Cells ; 41(5): 486-494, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29696935

ABSTRACT

Recently, we have reported that animals with telomerase reverse transcriptase (TERT) overexpression exhibit reduced social interaction, decreased preference for novel social interaction and poor nest-building behaviors symptoms that mirror those observed in human autism spectrum disorders (ASD). Overexpression of TERT also alters the excitatory/inhibitory (E/I) ratio in the medial prefrontal cortex. However, the effects of TERT overexpression on hippocampal-dependent learning and synaptic efficacy have not been investigated. In the present study, we employed electrophysiological approaches in combination with behavioral analysis to examine hippocampal function of TERT transgenic (TERT-tg) mice and FVB controls. We found that TERT overexpression results in enhanced hippocampal excitation with no changes in inhibition and significantly impairs long-term synaptic plasticity. Interestingly, the expression levels of phosphorylated CREB and phosphory-lated CaMKIIα were significantly decreased while the expression level of CaMKIIα was slightly increased in the hippocampus of TERT-overexpressing mice. Our observations highlight the importance of TERT in normal synaptic function and behavior and provide additional information on a novel animal model of ASD associated with TERT overexpression.


Subject(s)
Autism Spectrum Disorder/physiopathology , Disease Models, Animal , Neuronal Plasticity , Pyramidal Cells/physiology , Synaptic Transmission , Telomerase/physiology , Animals , Autism Spectrum Disorder/enzymology , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cyclic AMP Response Element-Binding Protein/biosynthesis , Cyclic AMP Response Element-Binding Protein/genetics , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gene Expression , Hippocampus/enzymology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurotoxins/pharmacology , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/enzymology , Recombinant Proteins/metabolism , Synaptic Transmission/drug effects , Telomerase/genetics , Tetrodotoxin/pharmacology
6.
Neuroscience ; 373: 207-217, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29406266

ABSTRACT

Isoprenoids and prenylated proteins regulate a variety of cellular functions, including neurite growth and synaptic plasticity. Importantly, they are implicated in the pathogenesis of several diseases, including Alzheimer's disease (AD). Recently, we have shown that two protein prenyltransferases, farnesyltransferase (FT) and geranylgeranyltransferase-1 (GGT), have differential effects in a mouse model of AD. Haplodeficiency of either FT or GGT attenuates amyloid-ß deposition and neuroinflammation but only reduction in FT rescues cognitive function. The current study aimed to elucidate the potential mechanisms that may account for the lack of cognitive benefit in GGT-haplodeficient mice, despite attenuated neuropathology. The results showed that the magnitude of long-term potentiation (LTP) was markedly suppressed in hippocampal slices from GGT-haplodeficient mice. Consistent with the synaptic dysfunction, there was a significant decrease in cortical spine density and cognitive function in GGT-haplodeficient mice. To further study the neuron-specific effects of GGT deficiency, we generated conditional forebrain neuron-specific GGT-knockout (GGTf/fCre+) mice using a Cre/LoxP system under the CAMKIIα promoter. We found that both the magnitude of hippocampal LTP and the dendritic spine density of cortical neurons were decreased in GGTf/fCre+ mice compared with GGTf/fCre- mice. Immunoblot analyses of cerebral lysate showed a significant reduction in cell membrane-associated (geranylgeranylated) Rac1 and RhoA but not (farnesylated) H-Ras, in GGTf/fCre+ mice, suggesting that insufficient geranylgeranylation of the Rho family of small GTPases may underlie the detrimental effects of GGT deficiency. These findings reinforce the critical role of GGT in maintaining spine structure and synaptic/cognitive function in development and in the mature brain.


Subject(s)
Alkyl and Aryl Transferases/deficiency , Brain/enzymology , Dendritic Spines/enzymology , Neuronal Plasticity/physiology , Alkyl and Aryl Transferases/genetics , Animals , Brain/pathology , Dendritic Spines/pathology , Excitatory Postsynaptic Potentials/physiology , Female , GTP Phosphohydrolases/metabolism , Male , Maze Learning/physiology , Mice, Transgenic , Pyramidal Cells/enzymology , Pyramidal Cells/pathology , Spatial Memory/physiology , Tissue Culture Techniques
7.
Cereb Cortex ; 28(4): 1329-1337, 2018 04 01.
Article in English | MEDLINE | ID: mdl-28334147

ABSTRACT

We described an extensive network of cortical pyramidal neurons in the human brain with abundant acetylcholinesterase (AChE) activity. Emergence of these neurons during childhood/adolescence, attainment of highest density in early adulthood, and virtual absence in other species led us to hypothesize involvement of AChE within these neurons in higher cortical functions. The current study quantified the density and staining intensity of these neurons using histochemical procedures. Few faintly stained AChE-positive cortical pyramidal neurons were observed in children/adolescents. These neurons attained their highest density and staining intensity in young adulthood. Compared with the young adult group, brains of cognitively normal elderly displayed no significant change in numerical density but a significant decrease in staining intensity of AChE-positive cortical pyramidal neurons. Brains of elderly above age 80 with unusually preserved memory performance (SuperAgers) showed significantly lower staining intensity and density of these neurons when compared with same-age peers. Conceivably, low levels of AChE activity could enhance the impact of acetylcholine on pyramidal neurons to counterbalance other involutional factors that mediate the decline of memory capacity during average aging. We cannot yet tell if elderly with superior memory capacity have constitutively low neuronal AChE levels or if this feature reflects adaptive neuroplasticity.


Subject(s)
Acetylcholinesterase/metabolism , Aging/physiology , Cerebral Cortex/cytology , Cognition/physiology , Pyramidal Cells/enzymology , Adult , Age Factors , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Neurofibrillary Tangles/physiology , Young Adult
8.
J Neurosci ; 38(3): 648-658, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29196323

ABSTRACT

Protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is one of four known kinases that respond to cellular stress by deactivating the eukaryotic initiation factor 2 α (eIF2α) or other signal transduction cascades. Recently, both eIF2α and its kinases were found to play a role in normal and pathological brain function. Here, we show that reduction of either the amount or the activity of PERK, specifically in the CA1 region of the hippocampus in young adult male mice, enhances neuronal excitability and improves cognitive function. In addition, this manipulation rescues the age-dependent cellular phenotype of reduced excitability and memory decline. Specifically, the reduction of PERK expression in the CA1 region of the hippocampus of middle-aged male mice using a viral vector rejuvenates hippocampal function and improves hippocampal-dependent learning. These results delineate a mechanism for behavior and neuronal aging and position PERK as a promising therapeutic target for age-dependent brain malfunction.SIGNIFICANCE STATEMENT We found that local reduced protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) expression or activity in the hippocampus enhances neuronal excitability and cognitive function in young normal mice, that old CA1 pyramidal cells have reduced excitability and increased PERK expression that can be rescued by reducing PERK expression in the hippocampus, and that reducing PERK expression in the hippocampus of middle-aged mice enhances hippocampal-dependent learning and memory and restores it to normal performance levels of young mice. These findings uncover an entirely new biological link among PERK, neuronal intrinsic properties, aging, and cognitive function. Moreover, our findings propose a new way to fight mild cognitive impairment and aging-related cognitive deterioration.


Subject(s)
Aging/physiology , Cognition/physiology , Hippocampus/enzymology , Hippocampus/metabolism , Memory/physiology , eIF-2 Kinase/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Cognition/drug effects , Cognitive Dysfunction/enzymology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Learning/drug effects , Learning/physiology , Male , Memory/drug effects , Mice , Pyramidal Cells/drug effects , Pyramidal Cells/enzymology
9.
Mol Cell Neurosci ; 85: 226-234, 2017 12.
Article in English | MEDLINE | ID: mdl-29107098

ABSTRACT

Stressor exposure induces neuronal remodeling in specific brain regions. Given the persistence of stress-related illnesses, key next steps in determining the contributions of neural structure to mental health are to identify cell types that fail to recover from stressor exposure and to identify "trigger points" and molecular underpinnings of stress-related neural degeneration. We evaluated dendrite arbor structure on hippocampal CA1 pyramidal neurons before, during, and following prolonged exposure to one key mediator of the stress response - corticosterone (cortisol in humans). Basal dendrite arbors progressively simplified during a 3-week exposure period, and failed to recover when corticosterone was withdrawn. Corticosterone exposure decreased levels of the dendrite stabilization factor Abl2/Arg nonreceptor tyrosine kinase and phosphorylation of its substrates p190RhoGAP and cortactin within 11days, suggesting that disruption of Arg-mediated signaling may trigger dendrite arbor atrophy and, potentially, behavioral abnormalities resulting from corticosterone exposure. To test this, we administered the novel, bioactive Arg kinase activator, 5-(1,3-diaryl-1H-pyrazol-4-yl)hydantoin, 5-[3-(4-fluorophenyl)-1-phenyl-1H-pyrazol-4-yl]-2,4-imidazolidinedione (DPH), in conjunction with corticosterone. We found that repeated treatment corrected CA1 arbor structure, otherwise simplified by corticosterone. DPH also corrected corticosterone-induced errors in a hippocampal-dependent reversal learning task and anhedonic-like behavior. Thus, pharmacological compounds that target cytoskeletal regulators, rather than classical neurotransmitter systems, may interfere with stress-associated cognitive decline and mental health concerns.


Subject(s)
Corticosterone/toxicity , Enzyme Activation/physiology , Protein-Tyrosine Kinases/metabolism , Pyramidal Cells/drug effects , Stress, Psychological/metabolism , Adrenal Cortex Hormones/toxicity , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/enzymology , Dendrites/drug effects , Dendrites/enzymology , Dendrites/pathology , Mice , Mice, Inbred C57BL , Pyramidal Cells/enzymology , Stress, Psychological/pathology
10.
Aging (Albany NY) ; 9(5): 1386-1403, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28522792

ABSTRACT

Defects in p21-activated kinase (PAK) lead to dendritic spine abnormalities and are sufficient to cause cognition impairment. The decrease in PAK in the brain of Alzheimer's disease (AD) patients is suspected to underlie synaptic and dendritic disturbances associated with its clinical expression, particularly with symptoms related to frontal cortex dysfunction. To investigate the role of PAK combined with Aß and tau pathologies (3xTg-AD mice) in the frontal cortex, we generated a transgenic model of AD with a deficit in PAK activity (3xTg-AD-dnPAK mice). PAK inactivation had no effect on Aß40 and Aß42 levels, but increased the phosphorylation ratio of tau in detergent-insoluble protein fractions in the frontal cortex of 18-month-old heterozygous 3xTg-AD mice. Morphometric analyses of layer II/III pyramidal neurons in the frontal cortex showed that 3xTg-AD-dnPAK neurons exhibited significant dendritic attrition, lower spine density and longer spines compared to NonTg and 3xTg-AD mice. Finally, behavioral assessments revealed that 3xTg-AD-dnPAK mice exhibited pronounced anxious traits and disturbances in social behaviors, reminiscent of fronto-dependent symptoms observed in AD. Our results substantiate a critical role for PAK in the genesis of neuronal abnormalities in the frontal cortex underlying the emergence of psychiatric-like symptoms in AD.


Subject(s)
Alzheimer Disease/enzymology , Behavior, Animal , Frontal Lobe/enzymology , Pyramidal Cells/enzymology , p21-Activated Kinases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Anxiety/enzymology , Anxiety/psychology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Disease Models, Animal , Exploratory Behavior , Frontal Lobe/pathology , Frontal Lobe/physiopathology , Genetic Predisposition to Disease , Interpersonal Relations , Locomotion , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/metabolism , Phenotype , Phosphorylation , Presenilin-1/genetics , Promoter Regions, Genetic , Pyramidal Cells/pathology , Synaptic Transmission , p21-Activated Kinases/genetics , tau Proteins/genetics , tau Proteins/metabolism
11.
Dev Neurobiol ; 77(10): 1161-1174, 2017 10.
Article in English | MEDLINE | ID: mdl-28388013

ABSTRACT

The formation and stabilization of new dendritic spines is a key component of the experience-dependent neural circuit plasticity that supports learning, but the molecular maturation of nascent spines remains largely unexplored. The PSD95-family of membrane-associated guanylate kinases (PSD-MAGUKs), most notably PSD95, has a demonstrated role in promoting spine stability. However, nascent spines contain low levels of PSD95, suggesting that other members of the PSD-MAGUK family might act to stabilize nascent spines in the early stages of spiny synapse formation. Here, we used GFP-fusion constructs to quantitatively define the molecular composition of new spines, focusing on the PSD-MAGUK family. We found that PSD95 levels in new spines were as low as those previously associated with rapid subsequent spine elimination, and new spines did not achieve mature levels of PSD95 until between 12 and 20 h following new spine identification. Surprisingly, we found that the PSD-MAGUKs PSD93, SAP97, and SAP102 were also substantially less enriched in new spines. However, they accumulated in new spines more quickly than PSD95: SAP102 enriched to mature levels within 3 h, SAP97 and PSD93 enriched gradually over the course of 6 h. Intriguingly, when we restricted our analysis to only those new spines that persisted, SAP97 was the only PSD-MAGUK already present at mature levels in persistent new spines when first identified. Our findings uncover a key structural difference between nascent and mature spines, and suggest a mechanism for the stabilization of nascent spines through the sequential arrival of PSD-MAGUKs. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1161-1174, 2017.


Subject(s)
Dendritic Spines/enzymology , Guanylate Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Female , Green Fluorescent Proteins , Hippocampus/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Microscopy, Confocal , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Pyramidal Cells/enzymology , Rats , Tissue Culture Techniques
12.
Neurochem Res ; 42(8): 2305-2313, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28349361

ABSTRACT

Glycogen synthase kinase 3ß (GSK-3ß) is a key downstream protein in the PI3K/Akt pathway. Phosphorylation of serine 9 of GSK-3ß (GSK-3ß activity inhibition) promotes cell survival. In this study, we examined changes in expressions of GSK-3ß and phosphorylation of GSK-3ß (p-GSK-3ß) in the gerbil hippocampal CA1 area after 5 min of transient cerebral ischemia. GSK-3ß immunoreactivity in the CA1 area was increased in pyramidal cells at 6 h after ischemia-reperfusion. It was decreased in CA1 pyramidal cells from 12 h after ischemia-reperfusion, and hardly detected in the CA1 pyramidal cells at 5 days after ischemia-reperfusion. p-GSK-3ß immunoreactivity was slightly decreased in CA1 pyramidal cells at 6 and 12 h after ischemia-reperfusion. It was significantly increased in these cells at 1 and 2 days after ischemia-reperfusion. Five days after ischemia-reperfusion, p-GSK-3ß immunoreactivity was hardly found in CA1 pyramidal cells. However, p-GSK-3ß immunoreactivity was strongly expressed in astrocytes primarily distributed in strata oriens and radiatum. In conclusion, GSK-3ß and p-GSK-3ß were significantly changed in pyramidal cells and/or astrocytes in the gerbil hippocampal CA1 area following 5 min of transient cerebral ischemia. This finding indicates that GSK-3ß and p-GSK-3ß are closely related to delayed neuronal death.


Subject(s)
Astrocytes/enzymology , Brain Ischemia/enzymology , CA1 Region, Hippocampal/enzymology , Gene Expression Regulation, Enzymologic , Glycogen Synthase Kinase 3 beta/biosynthesis , Pyramidal Cells/enzymology , Animals , Astrocytes/chemistry , Astrocytes/pathology , Avoidance Learning/physiology , Brain Ischemia/pathology , CA1 Region, Hippocampal/chemistry , CA1 Region, Hippocampal/pathology , Cell Death/physiology , Gerbillinae , Glycogen Synthase Kinase 3 beta/analysis , Glycogen Synthase Kinase 3 beta/genetics , Male , Pyramidal Cells/chemistry , Pyramidal Cells/pathology
13.
Neuropharmacology ; 117: 338-351, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28232062

ABSTRACT

Sodium-potassium ATPase (NaKA) is a plasma membrane enzyme responsible for influencing membrane physiology by direct electrogenic activity. It determines cellular excitability and synaptic neurotransmission, thus affecting learning and memory processes. A principle catalytic α subunit of NaKA has development-specific expression pattern. There are two α isoforms, α1 and α3, in adult brain neurons. Although NaKA is a housekeeping enzyme, the physiological differences between these two α isoforms in different brain regions have not been well explored. Endogenous cardiotonic steroids, including Marinobufagenin and Ouabain, control the cell homeostasis and cell functions via inhibiting NaKA. Here we employed selective inhibition of α1 and α3 NaKA isoforms by Marinobufagenin and Ouabain respectively, to measure the contribution of α subunits in cellular physiology of three distinct mouse brain regions. The results of the whole cell recording demonstrated that α1 isoform predominated in layer-5 pyramidal cells at rostral motor cortex, while α3 isoform governed the pyramidal neurons at hippocampal CA1 region and to a lesser extent the layer-5 pyramidal neurons of parietal cortex. Furthermore, selective α isoform inhibition induced differential effects on distinct physiological properties even within the same brain region. In addition, our results supported the existence of synergism between two NaKA α isoforms. To conclude, this systematic study of NaKA α isoforms demonstrated their broader roles in neuronal functioning in a region-specific manner.


Subject(s)
Brain/drug effects , Brain/enzymology , Pyramidal Cells/drug effects , Pyramidal Cells/enzymology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Bufanolides/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Mice, Inbred C57BL , Ouabain/pharmacology , Patch-Clamp Techniques , Tissue Culture Techniques
14.
J Biol Chem ; 292(14): 5645-5654, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28213516

ABSTRACT

Sialidase cleaves sialic acids on the extracellular cell surface as well as inside the cell and is necessary for normal long-term potentiation (LTP) at mossy fiber-CA3 pyramidal cell synapses and for hippocampus-dependent spatial memory. Here, we investigated in detail the role of sialidase in memory processing. Sialidase activity measured with 4-methylumbelliferyl-α-d-N-acetylneuraminic acid (4MU-Neu5Ac) or 5-bromo-4-chloroindol-3-yl-α-d-N-acetylneuraminic acid (X-Neu5Ac) and Fast Red Violet LB was increased by high-K+-induced membrane depolarization. Sialidase activity was also increased by chemical LTP induction with forskolin and activation of BDNF signaling, non-NMDA receptors, or NMDA receptors. The increase in sialidase activity with neural excitation appears to be caused not by secreted sialidase or by an increase in sialidase expression but by a change in the subcellular localization of sialidase. Astrocytes as well as neurons are also involved in the neural activity-dependent increase in sialidase activity. Sialidase activity visualized with a benzothiazolylphenol-based sialic acid derivative (BTP3-Neu5Ac), a highly sensitive histochemical imaging probe for sialidase activity, at the CA3 stratum lucidum of rat acute hippocampal slices was immediately increased in response to LTP-inducible high-frequency stimulation on a time scale of seconds. To obtain direct evidence for sialic acid removal on the extracellular cell surface during neural excitation, the extracellular free sialic acid level in the hippocampus was monitored using in vivo microdialysis. The free sialic acid level was increased by high-K+-induced membrane depolarization. Desialylation also occurred during hippocampus-dependent memory formation in a contextual fear-conditioning paradigm. Our results show that neural activity-dependent desialylation by sialidase may be involved in hippocampal memory processing.


Subject(s)
CA3 Region, Hippocampal/enzymology , Memory/physiology , Neuraminidase/metabolism , Pyramidal Cells/enzymology , Synaptic Transmission/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Female , Male , N-Acetylneuraminic Acid/metabolism , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism
15.
Physiol Behav ; 170: 115-123, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28017679

ABSTRACT

Although Metformin, a first-line antidiabetic drug, can ameliorate ischemia/reperfusion (I/R) induced brain damage, but how metformin benefits injured hippocampus and the mechanisms are still largely unknown. Therefore, the aim of this study was to investigate the neuroprotective mechanisms of metformin against ischemic brain damage induced by cerebral I/R and to explore whether the Akt-mediated down-regulation of the phosphorylation of JNK3 signaling pathway contributed to the protection provided by metformin. Transient global brain ischemia was induced by 4-vessel occlusion in adult male Sprague-Dawley rats. The open field tasks and Morris water maze were used to assess the effect of metformin on anxiety-like behavioral and cognitive impairment after I/R. Cresyl Violet staining was used to examine the survival of hippocampal CA1 pyramidal neurons. Immunoblotting was performed to measure the phosphorylation of Akt1, JNK3, c-Jun and the expression of cleaved caspase-3. Through ischemia/reperfusion (I/R) rat model, we found that metformin could attenuate the deficits of hippocampal related behaviors and inhibit cell apoptosis. The western blot data showed that metformin could promote the activation of Akt1 and reduce the phosphorylation of JNK3 and c-Jun as well as elevation of cleaved caspase-3 in I/R brains. PI3K inhibitor reversed all the protective effects, further indicating that metformin protect hippocampus from ischemic damage through PI3K/Akt1/JNK3/c-Jun signaling pathway.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/enzymology , Metformin/pharmacology , Neuroprotective Agents/pharmacology , Reperfusion Injury/drug therapy , Reperfusion Injury/enzymology , Animals , Anxiety/drug therapy , Anxiety/enzymology , Anxiety/etiology , Anxiety/pathology , Apoptosis/drug effects , Apoptosis/physiology , Brain Ischemia/complications , Brain Ischemia/pathology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/enzymology , CA1 Region, Hippocampal/pathology , Caspase 3/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Cognition Disorders/etiology , Cognition Disorders/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Mitogen-Activated Protein Kinase 10/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/enzymology , Pyramidal Cells/pathology , Rats, Sprague-Dawley , Reperfusion Injury/complications , Reperfusion Injury/pathology , Signal Transduction/drug effects
16.
J Alzheimers Dis ; 51(3): 783-91, 2016.
Article in English | MEDLINE | ID: mdl-26890755

ABSTRACT

Neurodegenerative diseases such as Alzheimer's disease (AD) are associated with alterations in epigenetic factors leading to cognitive decline. Histone deacetylase 3 (HDAC3) is a known critical epigenetic negative regulator of learning and memory. In this study, attenuation of long-term potentiation by amyloid-ß oligomer, and its reversal by specific HDAC3 inhibitor RGFP966, was performed in rat CA1 pyramidal neurons using whole cell voltage-clamp and field recording techniques. Our findings provide the first evidence that amyloid-ß oligomer-induced synaptic plasticity impairment can be prevented by inhibition of HDAC3 enzyme both at the single neuron as well as in a population of neurons, thus identifying HDAC3 as a potential target for ameliorating AD related plasticity impairments.


Subject(s)
Amyloid beta-Peptides/toxicity , Hippocampus/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Long-Term Potentiation/drug effects , Peptide Fragments/toxicity , Pyramidal Cells/drug effects , Acrylamides/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/enzymology , Long-Term Potentiation/physiology , Male , Patch-Clamp Techniques , Phenylenediamines/pharmacology , Pyramidal Cells/enzymology , Rats, Wistar , Tissue Culture Techniques
17.
Brain Struct Funct ; 221(4): 2147-62, 2016 05.
Article in English | MEDLINE | ID: mdl-25850901

ABSTRACT

Cytochrome b 5 reductase (Cb 5R) and cytochrome b 5 (Cb 5) form an enzymatic redox system that plays many roles in mammalian cells. In the last 15 years, it has been proposed that this system is involved in the recycling of ascorbate, a vital antioxidant molecule in the brain and that its deregulation can lead to the production of reactive oxygen species that play a major role in oxidative-induced neuronal death. In this work, we have performed a regional and cellular distribution study of the expression of this redox system in adult rat brain by anti-Cb 5R isoform 3 and anti-Cb 5 antibodies. We found high expression levels in cerebellar cortex, labeling heavily granule neurons and Purkinje cells, and in structures such as the fastigial, interposed and dentate cerebellar nuclei. A large part of Cb 5R isoform 3 in the cerebellum cortex was regionalized in close proximity to the lipid raft-like nanodomains, labeled with cholera toxin B, as we have shown by fluorescence resonance energy transfer imaging. In addition, vestibular, reticular and motor nuclei located at the brain stem level and pyramidal neurons of somatomotor areas of the brain cortex and of the hippocampus have been also found to display high expression levels of these proteins. All these results point out the enrichment of Cb 5R isoform 3/Cb 5 system in neuronal cells and structures of the cerebellum and brain stem whose functional impairment can account for neurological deficits reported in type II congenital methemoglobinemia, as well as in brain areas highly prone to undergo oxidative stress-induced neurodegeneration.


Subject(s)
Brain/enzymology , Cerebellum/enzymology , Cytochrome-B(5) Reductase/metabolism , Cytochromes b5/metabolism , Pyramidal Cells/enzymology , Animals , Brain Stem/enzymology , Hippocampus/enzymology , Isoenzymes/metabolism , Male , Membrane Microdomains/enzymology , Neocortex/enzymology , Neuroglia/enzymology , Rats , Rats, Wistar
18.
Bull Exp Biol Med ; 160(1): 96-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26608376

ABSTRACT

Adult rats were subjected to 7-day combined stress with stochastic changes of stressors of different modalities (noise, vibration, pulsating bright light) along with mobility restriction and elevated temperature in the chamber during stress exposures (daily 30-min sessions). Circulatory disorders, inhibition of endothelial NO-synthase expression in endothelial cells of the microcirculatory bed, perivascular edema, pronounced degenerative changes, and enhanced expression of inducible NO synthase in CA3 pyramidal neurons in the ventral hippocampus of stressed 12-month-old rats were observed. These findings can attest to the involvement NOdependent mechanisms and different contribution of NO synthase isoforms into the formation of hippocampal neuronal damage.


Subject(s)
CA3 Region, Hippocampal/enzymology , Nerve Tissue Proteins/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Pyramidal Cells/enzymology , Stress, Physiological , Animals , Animals, Outbred Strains , Brain Edema/enzymology , Brain Edema/etiology , Brain Edema/pathology , CA3 Region, Hippocampal/blood supply , CA3 Region, Hippocampal/ultrastructure , Endothelial Cells/enzymology , Enzyme Induction , Light/adverse effects , Male , Microcirculation , Nerve Degeneration/enzymology , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Tissue Proteins/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III/genetics , Noise/adverse effects , Pyramidal Cells/ultrastructure , Rats , Restraint, Physical/adverse effects , Temperature , Vibration/adverse effects
19.
J Neurosci ; 35(29): 10510-22, 2015 Jul 22.
Article in English | MEDLINE | ID: mdl-26203146

ABSTRACT

Neuronal death caused by excessive excitatory signaling, excitotoxicity, plays a central role in neurodegenerative disorders. The mechanisms regulating this process, however, are still incompletely understood. Here we show that the coated vesicle-associated kinase SCYL2/CVAK104 plays a critical role for the normal functioning of the nervous system and for suppressing excitotoxicity in the developing hippocampus. Targeted disruption of Scyl2 in mice caused perinatal lethality in the vast majority of newborn mice and severe sensory-motor deficits in mice that survived to adulthood. Consistent with a neurogenic origin of these phenotypes, neuron-specific deletion of Scyl2 also caused perinatal lethality in the majority of newborn mice and severe neurological defects in adult mice. The neurological deficits in these mice were associated with the degeneration of several neuronal populations, most notably CA3 pyramidal neurons of the hippocampus, which we analyzed in more detail. The loss of CA3 neurons occurred during the functional maturation of the hippocampus and was the result of a BAX-dependent apoptotic process. Excessive excitatory signaling was present at the onset of degeneration, and inhibition of excitatory signaling prevented the degeneration of CA3 neurons. Biochemical fractionation reveals that Scyl2-deficient mice have an altered composition of excitatory receptors at synapses. Our findings demonstrate an essential role for SCYL2 in regulating neuronal function and survival and suggest a role for SCYL2 in regulating excitatory signaling in the developing brain. Significance statement: Here we examine the in vivo function of SCYL2, an evolutionarily conserved and ubiquitously expressed protein pseudokinase thought to regulate protein trafficking along the secretory pathway, and demonstrate its importance for the normal functioning of the nervous system and for suppressing excitatory signaling in the developing brain. Together with recent studies demonstrating a role of SCYL1 in preventing motor neuron degeneration, our findings clearly establish the SCY1-like family of protein pseudokinases as key regulators of neuronal function and survival.


Subject(s)
CA3 Region, Hippocampal/enzymology , Nerve Degeneration/enzymology , Neurogenesis/physiology , Protein Serine-Threonine Kinases/metabolism , Pyramidal Cells/enzymology , Animals , Blotting, Western , Cell Death/physiology , Chromatography, Liquid , Electrophysiology , Excitatory Postsynaptic Potentials/physiology , Fluorescent Antibody Technique , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Confocal , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry
20.
Mol Med Rep ; 12(2): 2555-62, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25955690

ABSTRACT

The thioredoxin (Trx) and peroxiredoxin (Prx) redox system is associated with neuronal damage and neuroprotective effects via the regulation of oxidative stress in brain ischemia. In the present study, ischemia-induced changes in the protein expression levels of Trx2 and Prx3 in the stratum pyramidale (SP) of the hippocampal CA1 region were investigated in adult and aged gerbils, subjected to 5 min of transient global cerebral ischemia, using immunohistochemistry and western blot analysis. In the adult ischemia-group, minimal Trx2 immunoreactivity was detected in the SP 2 days after ischemia-reperfusion. In the aged animals, the Trx2 immunoreactivity in the sham-group was marginally lower compared with that in the adult sham-group. In the aged ischemia-group, Trx2 immunoreactivity in the SP was significantly higher 1, 2 and 4 days post-ischemia, compared with that in the adult ischemia-group and, in the 5 days post-ischemia group, Trx2 immunoreactivity was significantly decreased in the SP. Prx3 immunoreactivity in the SP of the adult ischemia-group was significantly decreased from 4 days after ischemia-reperfusion. In the aged animals, Prx3 immunoreactivity in the sham-group was also marginally lower compared with that in the adult sham-group. Prx3 immunoreactivity in the aged ischemia-group was also significantly higher 1, 2 and 4 days post-ischemia, compared with the adult ischemia-group; however, the Prx3 immunoreactivity was significantly decreased 5 days post-ischemia. The western blot analyses revealed that the pattern of changes in the protein levels of Trx2 and Prx3 in the adult and aged hippocampal CA1 region following ischemic damage were similar to the results obtained in the immunohistochemical data. These findings indicated that cerebral ischemia lead to different protein expression levels of Trx2 and Prx3 in the hippocampal CA1 region between adult and aged gerbils, and these differences may be associated with more delayed neuronal death in the aged gerbil hippocampus following transient global cerebral ischemia.


Subject(s)
Aging/metabolism , CA1 Region, Hippocampal/enzymology , Ischemic Attack, Transient/genetics , Peroxiredoxin III/genetics , Pyramidal Cells/enzymology , Thioredoxins/genetics , Aging/pathology , Animals , CA1 Region, Hippocampal/pathology , Gene Expression Regulation , Gerbillinae , Immunohistochemistry , Ischemic Attack, Transient/enzymology , Ischemic Attack, Transient/pathology , Male , Oxidative Stress , Peroxiredoxin III/metabolism , Pyramidal Cells/pathology , Signal Transduction , Thioredoxins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...