Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
1.
Int J Sports Med ; 41(13): 951-961, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32643775

ABSTRACT

Exercise has been argued to improve cognitive function in both humans and rodents. Angiogenesis significantly contributes to brain health, including cognition. The hippocampus is a crucial brain region for cognitive function. However, studies quantifying the capillary changes in the hippocampus after running exercise are lacking. Moreover, the molecular details underlying the effects of running exercise remain poorly understood. We show that endogenous nitric oxide contributes to the beneficial effects of running exercise on cognition and hippocampal capillaries. Four weeks of running exercise significantly improved spatial memory ability and increased the number of capillaries in the cornu ammonis 1 subfield and dentate gyrus of Sprague-Dawley rats. Running exercise also significantly increased nitric oxide synthase activity and nitric oxide content in the rat hippocampus. After blocking the synthesis of endogenous nitric oxide by lateral ventricular injection of NG-nitro-L-arginine methyl ester, a nonspecific nitric oxide synthase inhibitor, the protective effect of running exercise on spatial memory was eliminated. The protective effect of running exercise on angiogenesis in the cornu ammonis 1 subfield and dentate gyrus of rats was also absent after nitric oxide synthase inhibition. Therefore, during running excise, endogenous nitric oxide may contribute to regulating spatial memory ability and angiogenesis in cornu ammonis 1 subfield and dentate gyrus of the hippocampus.


Subject(s)
CA1 Region, Hippocampal/blood supply , Capillaries/physiology , Dentate Gyrus/blood supply , Neovascularization, Physiologic , Nitric Oxide/physiology , Physical Conditioning, Animal/physiology , Spatial Memory/physiology , Animals , CA1 Region, Hippocampal/enzymology , Dentate Gyrus/enzymology , Male , Maze Learning/physiology , Nitric Oxide Synthase/metabolism , Rats, Sprague-Dawley , Running/physiology
2.
Hippocampus ; 29(7): 569-578, 2019 07.
Article in English | MEDLINE | ID: mdl-30387892

ABSTRACT

The hippocampus is generally considered as a brain center for learning and memory. We have recently established an electroporation-mediated gene transfer method to investigate the development of neonatal dentate granule cells in vivo. Using this new technique, we introduced knockdown vectors against Rac1 small GTPase into precursors for dentate granule cells at postnatal day 0. After 21 days, Rac1-deficient cells were frequently mispositioned between the granule cell layer (GCL) and hilus. About 60% of these mislocalized cells expressed a dentate granule cell marker, Prox1. Both the dendritic spine density and the ratio of mature spine were reduced when Rac1 was silenced. Notably, the deficient cells have immature thin processes during migrating in the early neonatal period. Knockdown of another Rac isoform, Rac3, also resulted in mislocalization of neonatally born dentate granule cells. In addition, knockdown of Cdc42, another Rho family protein, also caused mislocalization of the cell, although the effects were moderate compared to Rac1 and 3. Despite the ectopic localization, Rac3- or Cdc42-disrupted mispositioned cells expressed Prox1. These results indicate that Rho signaling pathways differentially regulate the proper localization and differentiation of dentate granule cells.


Subject(s)
Dentate Gyrus/enzymology , Dentate Gyrus/growth & development , Neuropeptides/metabolism , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cell Movement , Dentate Gyrus/cytology , Gene Knockdown Techniques , Gene Transfer Techniques , Homeodomain Proteins/metabolism , Mice , Mice, Inbred ICR , Neurogenesis , Neuropeptides/deficiency , Neuropeptides/genetics , RNA Interference , Signal Transduction , Tumor Suppressor Proteins/metabolism , cdc42 GTP-Binding Protein/deficiency , cdc42 GTP-Binding Protein/genetics , rac GTP-Binding Proteins/deficiency , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein/deficiency , rac1 GTP-Binding Protein/genetics
3.
J Neurochem ; 144(1): 58-67, 2018 01.
Article in English | MEDLINE | ID: mdl-29063591

ABSTRACT

Ghrelin is an appetite-stimulating peptide. Serine 3 on ghrelin must be acylated by octanoate via the enzyme ghrelin-O-acyltransferase (GOAT) for the peptide to bind and activate the cognate receptor, growth hormone secretagogue receptor type 1a (GHSR1a). Interest in GHSR1a increased dramatically when GHSR1a mRNA was demonstrated to be widespread in the brain, including the cortex and hippocampus, indicating that it has multifaceted functions beyond the regulation of metabolism. However, the source of octanoylated ghrelin for GHSR1a in the brain, outside of the hypothalamus, is not well understood. Here, we report the presence of GOAT and its ability to acylate non-octanoylated ghrelin in the hippocampus. GOAT immunoreactivity is aggregated at the base of the dentate granule cell layer in the rat and wild-type mouse. This immunoreactivity was not affected by the pharmacological inhibition of GHSR1a or the metabolic state-dependent fluctuation of systemic ghrelin levels. However, it was absent in the GHSR1a knockout mouse hippocampus, pointing the possibility that the expression of GHSR1a may be a prerequisite for the production of GOAT. Application of fluorescein isothiocyanate (FITC)-conjugated non-octanoylated ghrelin in live hippocampal slice culture (but not in fixed culture or in the presence of GOAT inhibitors) mimicked the binding profile of FITC-conjugated octanoylated ghrelin, suggesting that extracellularly applied non-octanoylated ghrelin was acylated by endogenous GOAT in the live hippocampus while GOAT being mobilized out of neurons. Our results will advance the understanding for the role of endogenous GOAT in the hippocampus and facilitate the search for the source of ghrelin that is intrinsic to the brain.


Subject(s)
Acyltransferases/metabolism , Dentate Gyrus/enzymology , Ghrelin/metabolism , Acylation , Animals , Caprylates/metabolism , Female , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Ghrelin/pharmacology , Male , Membrane Proteins , Mice , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/deficiency , Receptors, Ghrelin/physiology
4.
Nitric Oxide ; 81: 67-74, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29074292

ABSTRACT

INTRODUCTION: We previously demonstrated that cyclic AMP-dependent protein kinase (PKA) phosphorylates neuronal nitric oxide synthase (nNOS) at Ser1412 in the hippocampal dentate gyrus after forebrain ischemia; this phosphorylation event activates NOS activity and might contribute to depression after cerebral ischemia. In this study, we revealed chronological and topographical changes in the phosphorylation of nNOS at Ser1412 immediately after subarachnoid hemorrhage (SAH). METHODS: In a rat single-hemorrhage model of SAH, the hippocampus and adjacent cortex were collected up to 24 h after SAH. Samples from rats that were not injected with autologous blood were used as controls. NOS was partially purified from crude samples via an ADP-agarose gel. Levels of nNOS, nNOS phosphorylated at Ser1412 (p-nNOS), PKA, and p-PKA at Thr197 were studied in the rat hippocampus and cortex using Western blot analyses and immunohistochemistry. RESULTS: According to the Western blot analysis, levels of p-nNOS at Ser1412 were significantly increased in the hippocampus, but not in the cortex, between 1 and 3 h after SAH. Immunohistochemistry revealed the phosphorylation of nNOS at Ser1412 and PKA at Thr197 in the dentate gyrus, but not in the CA1 area, 1 h after SAH. An injection of saline instead of blood also significantly increased levels of p-nNOS at Ser1412 in the hippocampus 1 h after the injection. CONCLUSIONS: An immediate increase in intracranial pressure (ICP) might induce transient cerebral ischemia and promote the PKA-mediated phosphorylation of nNOS at Ser1412 in the dentate gyrus. This signal transduction pathway induces the excessive production of nitric oxide (NO) and might be involved in cognitive dysfunction after SAH.


Subject(s)
Dentate Gyrus/enzymology , Nitric Oxide Synthase Type I/metabolism , Subarachnoid Hemorrhage/enzymology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Dentate Gyrus/metabolism , Male , Neurons/enzymology , Neurons/pathology , Phosphorylation , Rats, Sprague-Dawley , Serine/metabolism , Subarachnoid Hemorrhage/metabolism , Threonine/metabolism
5.
Pharmacology ; 102(1-2): 42-52, 2018.
Article in English | MEDLINE | ID: mdl-29843142

ABSTRACT

BACKGROUND/AIMS: Decreasing levels of aromatase and seladin-1 could be one of the molecular mechanisms of Alzheimer's disease (AD). Aromatase is an enzyme that catalyzes estrogen biosynthesis from androgen precursors, and seladin-1 is an enzyme that converts desmosterol to cholesterol, which is the precursor of all hormones. Verifying the potential relationship between these proteins and accordingly determining new therapeutic targets constitute the aims of this study. METHODS: Changes in protein levels were compared in vitro in aromatase and seladin-1 inhibitor-administered human neuroblastoma (SH-SY5Y) cells in vivo in intracerebroventricular (icv) aromatase or seladin-1 inhibitor-administered rats, as well as in transgenic AD mice in which the genes encoding these proteins were knocked out. RESULTS AND CONCLUSIONS: In the cell cultures, we observed that seladin-1 protein levels increased after aromatase enzyme inhibition. The hippocampal aromatase protein levels decreased following chronic seladin-1 inhibition in icv inhibitor-administered rats; however, the aromatase levels in the dentate gyrus of seladin-1 knockout (SelKO) AD male mice increased. These findings indicate a partial relationship between these proteins and their roles in AD pathology.


Subject(s)
Alzheimer Disease/enzymology , Aromatase/metabolism , Hippocampus/enzymology , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Androstenes/pharmacology , Animals , Aromatase/genetics , Aromatase Inhibitors/administration & dosage , Aromatase Inhibitors/pharmacology , Cells, Cultured , Dentate Gyrus/enzymology , Female , Humans , Infusions, Intraventricular , Letrozole , Male , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/enzymology , Nitriles/pharmacology , Oxidoreductases Acting on CH-CH Group Donors/genetics , Rats , Triazoles/pharmacology
6.
Cell Mol Neurobiol ; 37(5): 869-877, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27734244

ABSTRACT

Some anticonvulsant drugs are associated with cognitive ability in patients; Topiramate (TPM) is well known as an effective anticonvulsant agent applied in clinical settings. However, the effect of TPM on the cognitive function is rarely studied. In this study, we aimed to observe the effects of TPM on cell proliferation and neuronal differentiation in the dentate gyrus (DG) of the D-galactose-induced aging mice by Ki-67 and doublecortin (DCX) immunohistochemistry. The study is divided into four groups including control, D-galactose-treated group, 25 and 50 mg/kg TPM-treated plus D-galactose-treated groups. We found, 50 mg/kg (not 25 mg/kg) TPM treatment significantly increased the numbers of Ki-67+ cells and DCX immunoreactivity, and improved neuroblast injury induced by D-galactose treatment. In addition, we also found that decreased immunoreactivities and protein levels of antioxidants including superoxide dismutase and catalase induced by D-galactose treatment were significantly recovered by 50 mg/kg TPM treatment in the mice hippocampal DG (P < 0.05). In conclusion, our present results indicate that TPM can ameliorate neuroblast damage and promote cell proliferation and neuroblast differentiation in the hippocampal DG via increasing SODs and catalase levels in the D-galactose mice.


Subject(s)
Aging/physiology , Antioxidants/pharmacology , Cell Differentiation/drug effects , Dentate Gyrus/cytology , Fructose/analogs & derivatives , Galactose/adverse effects , Neurons/cytology , Animals , Catalase/metabolism , Cell Proliferation/drug effects , Dentate Gyrus/drug effects , Dentate Gyrus/enzymology , Doublecortin Domain Proteins , Doublecortin Protein , Fructose/pharmacology , Mice , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Superoxide Dismutase/metabolism , Topiramate
7.
Proc Natl Acad Sci U S A ; 111(37): 13541-6, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25161285

ABSTRACT

The maintenance of the resident adult neural stem/progenitor cell (NSPC) pool depends on the precise balance of proliferation, differentiation, and maintenance of the undifferentiated state. Identifying the mechanisms that regulate this balance in adult hippocampal NSPCs can provide insight into basic stem cell self-renewal principles important for tissue homeostasis and preventing tumor formation. Pharmacological inhibition of histone deacetylases (HDACs), a class of histone-modifying enzymes, have promising effects in cancer cells, yet the specific roles of individual HDACs in stem cell proliferation is unclear. Here using conditional KO (cKO) mice and in vitro cell culture, we show that histone deacetylase 3 (HDAC3) is required for the proliferation of adult NSPCs. Detailed cell cycle analysis of NSPCs from Hdac3 cKO mice reveals a defect in cell cycle progression through the gap 2/mitosis (G2/M) but not the S phase. Moreover, HDAC3 controls G2/M phase progression mainly through posttranslational stabilization of the G2/M cyclin-dependent kinase 1 (CDK1). These results demonstrate that HDAC3 plays a critical role in NSPC proliferation and suggest that strategies aimed at pharmacological modulation of HDAC3 may be beneficial for tissue regeneration and controlling tumor cell growth.


Subject(s)
Adult Stem Cells/cytology , CDC2 Protein Kinase/metabolism , G2 Phase , Histone Deacetylases/metabolism , Mitosis , Neural Stem Cells/cytology , Adult Stem Cells/enzymology , Aging/physiology , Animals , Cell Proliferation , Dentate Gyrus/cytology , Dentate Gyrus/enzymology , Down-Regulation , Gene Deletion , Mice, Knockout , Neural Stem Cells/enzymology , Neurogenesis , Neurons/cytology , Neurons/enzymology , Proteolysis
8.
J Neurosci ; 35(17): 6836-48, 2015 Apr 29.
Article in English | MEDLINE | ID: mdl-25926459

ABSTRACT

The ERK/MAPK pathway is an important developmental signaling pathway. Mutations in upstream elements of this pathway result in neuro-cardio-facial cutaneous (NCFC) syndromes, which are typified by impaired neurocognitive abilities that are reliant upon hippocampal function. The role of ERK signaling during hippocampal development has not been examined and may provide critical insight into the cause of hippocampal dysfunction in NCFC syndromes. In this study, we have generated ERK1 and conditional ERK2 compound knock-out mice to determine the role of ERK signaling during development of the hippocampal dentate gyrus. We found that loss of both ERK1 and ERK2 resulted in 60% fewer granule cells and near complete absence of neural progenitor pools in the postnatal dentate gyrus. Loss of ERK1/2 impaired maintenance of neural progenitors as they migrate from the dentate ventricular zone to the dentate gyrus proper, resulting in premature depletion of neural progenitor cells beginning at E16.5, which prevented generation of granule cells later in development. Finally, loss of ERK2 alone does not impair development of the dentate gyrus as animals expressing only ERK1 developed a normal hippocampus. These findings establish that ERK signaling regulates maintenance of progenitor cells required for development of the dentate gyrus.


Subject(s)
Dentate Gyrus , Feedback, Physiological/physiology , Gene Expression Regulation, Developmental/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Stem Cells/physiology , Animals , Animals, Newborn , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Dentate Gyrus/embryology , Dentate Gyrus/enzymology , Dentate Gyrus/growth & development , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental/genetics , Lateral Ventricles/cytology , Lateral Ventricles/embryology , Lateral Ventricles/growth & development , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neurons/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism
9.
Hippocampus ; 26(4): 502-16, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26443682

ABSTRACT

Anxiety disorders are presumably associated with negative memory. Psychological therapies are widely used to treat this mental deficit in human beings based on the view that positive memory competes with negative memory and relieves anxiety status. Cellular and molecular processes underlying psychological therapies remain elusive. Therefore, we have investigated its mechanisms based on a mouse model in which food reward at one open-arm of the elevated plus-maze was used for training mice to form reward memory and challenge the open arms. Mice with the reward training showed increased entries and stay time in reward open-arm versus neutral open-arm as well as in open-arms versus closed-arms. Accompanying with reward memory formation and anxiety relief, glutamatergic synaptic transmission in dentate gyrus in vivo and dendritic spines in granule cells became upregulated. This synaptic up-regulation was accompanied by the expression of more protein kinase C (PKC) in the dendritic spines. The inhibition of PKC by chelerythrine impaired the formation of reward memory, the relief of anxiety-related behavior and the up-regulation of glutamate synapses. Our results suggest that reward-induced positive memory relieves mouse anxiety-related behavior by strengthening synaptic efficacy and PKC in the hippocampus, which imply the underlying cellular and molecular processes involved in the beneficial effects of psychological therapies treating anxiety disorders.


Subject(s)
Anxiety Disorders/therapy , Dentate Gyrus/enzymology , Memory/physiology , Protein Kinase C/metabolism , Reward , Synapses/enzymology , Animals , Anxiety Disorders/enzymology , Anxiety Disorders/pathology , Anxiety Disorders/psychology , Benzophenanthridines/pharmacology , Dendritic Spines/drug effects , Dendritic Spines/enzymology , Dendritic Spines/pathology , Dentate Gyrus/drug effects , Disease Models, Animal , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Mice, Inbred DBA , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Synapses/drug effects , Synapses/pathology , Up-Regulation
10.
Nutr Neurosci ; 19(6): 247-59, 2016 Jul.
Article in English | MEDLINE | ID: mdl-25923962

ABSTRACT

OBJECTIVES: This study aimed to investigate the beneficial effects of Cheonggukjang (CGK) manufactured by mixed culture of Bacillus subtilis MC31 and Lactobacillus sakei 383 on neurotoxic damages. METHODS: The specific aspects of brain functions were measured in Institute for Cancer Research (ICR) mice that had been pretreated for 4 weeks with three difference doses of CGK before trimethyltin (TMT) treatment. RESULTS: The short- and long-term memory loss induced by TMT treatment was significantly improved in the CGK-pretreated group in a dose-dependent manner. The number of dead cells in the granule cell layer of the dentate gyrus was decreased in the TMT/CGK-cotreated group relative to the TMT/vehicle-treated group, whereas significant suppression of acetylcholinesterase (AChE) activity was observed in the same group. Additionally, a dose-dependent increase in nerve growth factor (NGF) concentration, activation of the NGF receptor signaling pathway including the TrkA high affinity receptor and p75(NTR) low affinity receptor, and decline in Bax/Bcl-2 level was measured in all TMT/CGK-treated groups, although a decrease in the active form of caspase-3 was observed in the TMT/H-CGK-treated group. Furthermore, superoxide dismutase (SOD) activity was enhanced in the TMT/CGK-treated group, whereas the level of malondialdehyde (MDA), a marker of lipid peroxidation, was 43-58% lower in the TMT/CGK-treated group than the TMT/vehicle-treated group. DISCUSSION: These results demonstrate that CGK fermented by mixed culture of B. subtilis and L. sakei could exert a wide range of beneficial activities for neurodegenerative diseases, including Alzheimer, Parkinson, and Huntington disease.


Subject(s)
Bacillus subtilis/metabolism , Cognition Disorders/prevention & control , Dietary Supplements , Latilactobacillus sakei/metabolism , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Soy Foods/analysis , Animals , Biomarkers/metabolism , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Dentate Gyrus/drug effects , Dentate Gyrus/enzymology , Dentate Gyrus/pathology , Dietary Supplements/analysis , Fermentation , Functional Food/analysis , Functional Food/microbiology , Heavy Metal Poisoning, Nervous System/physiopathology , Lipid Peroxidation/drug effects , Male , Memory Disorders/etiology , Memory Disorders/metabolism , Memory Disorders/pathology , Memory Disorders/prevention & control , Mice , Mice, Inbred ICR , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neuroprotective Agents/chemistry , Plant Extracts/chemistry , Specific Pathogen-Free Organisms , Trimethyltin Compounds/toxicity
11.
Hum Mol Genet ; 22(7): 1300-15, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23257288

ABSTRACT

Adult neurogenesis, the generation of new neurons during the adulthood, is a process controlled by several kinases and phosphatases among which GSK3ß exerts important functions. This protein is particularly abundant in the central nervous system, and its activity deregulation is believed to play a key role in chronic disorders such as Alzheimer's disease. Previously, we reported that in vivo overexpression of GSK3ß (Tet/GSK3ß mice) causes alterations in adult neurogenesis, leading to a depletion of the neurogenic niches. Here, we have further characterized those alterations, finding a delay in the switching-off of doublecortin marker as well as changes in the survival and death rates of immature precursors and a decrease in the total number of mature neurons. Besides, we have highlighted the importance of the inflammatory environment, identifying eotaxin as a possible modulator of the detrimental effects on adult neurogenesis. Taking advantage of the conditional system, we have also explored whether these negative consequences of increasing GSK3 activity are susceptible to revert after doxycycline treatment. We show that transgene shutdown in symptomatic mice reverts microgliosis, abnormal eotaxin levels as well as the aforementioned alterations concerning immature neurons. Unexpectedly, the decrease in the number of mature neurons and neuronal precursor cells of the subgranular zone of Tet/GSK3ß mice could not be reverted. Thus, alterations in adult neurogenesis and likely in neurodegenerative disorders can be restored in part, although neurogenic niche depletion represents a non-reversible damage persisting during lifetime with a remarkable impact in adult mature neurons.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Neurogenesis , Animals , Biomarkers/metabolism , Cell Survival , Chemokine CCL11/metabolism , DNA-Binding Proteins , Dentate Gyrus/cytology , Dentate Gyrus/enzymology , Doublecortin Domain Proteins , Enzyme Induction , Genes, Reporter , Glial Fibrillary Acidic Protein/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Intermediate Filament Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Neural Stem Cells/physiology , Neurons/enzymology , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Stem Cell Niche , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
12.
J Neurosci ; 32(19): 6444-55, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22573667

ABSTRACT

Although there is evidence suggesting that adult neurogenesis may contribute to hippocampus-dependent memory, signaling mechanisms responsible for adult hippocampal neurogenesis are not well characterized. Here we report that ERK5 mitogen-activated protein kinase is specifically expressed in the neurogenic regions of the adult mouse brain. The inducible and conditional knock-out (icKO) of erk5 specifically in neural progenitors of the adult mouse brain attenuated adult hippocampal neurogenesis. It also caused deficits in several forms of hippocampus-dependent memory, including contextual fear conditioning generated by a weak footshock. The ERK5 icKO mice were also deficient in contextual fear extinction and reversal of Morris water maze spatial learning and memory, suggesting that adult neurogenesis plays an important role in hippocampus-dependent learning flexibility. Furthermore, our data suggest a critical role for ERK5-mediated adult neurogenesis in pattern separation, a form of dentate gyrus-dependent spatial learning and memory. Moreover, ERK5 icKO mice have no memory 21 d after training in the passive avoidance test, suggesting a pivotal role for adult hippocampal neurogenesis in the expression of remote memory. Together, our results implicate ERK5 as a novel signaling molecule regulating adult neurogenesis and provide strong evidence that adult neurogenesis is critical for several forms of hippocampus-dependent memory formation, including fear extinction, and for the expression of remote memory.


Subject(s)
Extinction, Psychological/physiology , Fear/physiology , Gene Deletion , Memory, Long-Term/physiology , Mitogen-Activated Protein Kinase 7/deficiency , Mitogen-Activated Protein Kinase 7/genetics , Neural Inhibition/genetics , Neurogenesis/physiology , Aging/genetics , Animals , Dentate Gyrus/enzymology , Dentate Gyrus/physiology , Gene Targeting/methods , Male , Mice , Mice, Knockout , Neurogenesis/genetics , Random Allocation , Signal Transduction/genetics
13.
J Neurosci ; 32(40): 13987-99, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23035106

ABSTRACT

Traf2 and NcK interacting kinase (TNiK) contains serine-threonine kinase and scaffold domains and has been implicated in cell proliferation and glutamate receptor regulation in vitro. Here we report its role in vivo using mice carrying a knock-out mutation. TNiK binds protein complexes in the synapse linking it to the NMDA receptor (NMDAR) via AKAP9. NMDAR and metabotropic receptors bidirectionally regulate TNiK phosphorylation and TNiK is required for AMPA expression and synaptic function. TNiK also organizes nuclear complexes and in the absence of TNiK, there was a marked elevation in GSK3ß and phosphorylation levels of its cognate phosphorylation sites on NeuroD1 with alterations in Wnt pathway signaling. We observed impairments in dentate gyrus neurogenesis in TNiK knock-out mice and cognitive testing using the touchscreen apparatus revealed impairments in pattern separation on a test of spatial discrimination. Object-location paired associate learning, which is dependent on glutamatergic signaling, was also impaired. Additionally, TNiK knock-out mice displayed hyperlocomotor behavior that could be rapidly reversed by GSK3ß inhibitors, indicating the potential for pharmacological rescue of a behavioral phenotype. These data establish TNiK as a critical regulator of cognitive functions and suggest it may play a regulatory role in diseases impacting on its interacting proteins and complexes.


Subject(s)
Association Learning/physiology , Cognition Disorders/enzymology , Dentate Gyrus/enzymology , Discrimination Learning/physiology , Nerve Tissue Proteins/physiology , Post-Synaptic Density/enzymology , Protein Serine-Threonine Kinases/physiology , Signal Detection, Psychological/physiology , Space Perception/physiology , Animals , Cell Nucleus/enzymology , Cognition Disorders/physiopathology , Dentate Gyrus/pathology , Glutamic Acid/physiology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Mice , Mice, Inbred C57BL , Mice, Knockout , Miniature Postsynaptic Potentials/physiology , Nerve Tissue Proteins/deficiency , Neurogenesis/physiology , Phenotype , Phosphorylation , Post-Synaptic Density/physiology , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/physiology
14.
J Proteome Res ; 12(12): 5422-35, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24200092

ABSTRACT

The effect of aging on hippocampus is often confounded by diseases that commonly occur in the elderly. In this research, functional proteomics was used to characterize age-related changes in energy metabolism of different neuronal pathways within the hippocampus of Wistar rats aged 2, 6, 12, 18, and 24 months. The "large" synaptosomes, derived from glutamatergic mossy fiber endings connecting granule cells of dentate gyrus with apical dendrites of CA3 pyramidal cells, and the "small" synaptosomes, derived from the cholinergic small nerve endings of septo-hippocampal fibers, whose projections reach CA1 pyramidal cells, were isolated. Because most brain disorders are associated with bioenergetic changes, the maximum rate (V(max)) of selected enzymes of glycolysis, Krebs cycle, glutamate and amino acids metabolism, and acetylcholine catabolism were evaluated. The results show that "large" and "small" synaptosomes possess specific and independent metabolic features coherently with the selective vulnerability of the respective hippocampal subfields to Alzheimer's disease and cerebral ischemia. This study represents a reliable model to study in vivo (i) the physiopathological molecular mechanisms of some brain diseases dependent on energy metabolism, (ii) the responsiveness to noxious stimuli, and (iii) the effects of drugs, discriminating their action sites at subcellular level.


Subject(s)
Aging/metabolism , CA1 Region, Hippocampal/enzymology , Dentate Gyrus/enzymology , Pyramidal Cells/enzymology , Synaptosomes/enzymology , Acetylcholine/metabolism , Aging/pathology , Animals , CA1 Region, Hippocampal/pathology , Citric Acid Cycle , Dentate Gyrus/pathology , Glutamic Acid/metabolism , Glycolysis , Humans , Kinetics , Male , Neurons/enzymology , Neurons/pathology , Proteomics , Pyramidal Cells/pathology , Rats , Rats, Wistar , Synaptosomes/pathology
15.
Neurobiol Learn Mem ; 105: 81-92, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23871742

ABSTRACT

Adult neurogenesis occurs in two discrete regions of the adult mammalian brain, the subgranular zone (SGZ) of the dentate gyrus (DG) and the subventricular zone (SVZ) along the lateral ventricles. Signaling mechanisms regulating adult neurogenesis in the SGZ are currently an active area of investigation. Adult-born neurons in the DG functionally integrate into the hippocampal circuitry and form functional synapses, suggesting a role for these neurons in hippocampus-dependent memory formation. Although results from earlier behavioral studies addressing this issue were inconsistent, recent advances in conditional gene targeting technology, viral injection and optogenetic approaches have provided convincing evidence supporting a role for adult-born neurons in the more challenging forms of hippocampus-dependent learning and memory. Here, we briefly summarize these recent studies with a focus on extra signal-regulated kinase (ERK) 5, a MAP kinase whose expression in the adult brain is restricted to the neurogenic regions including the SGZ and SVZ. We review evidence identifying ERK5 as a novel endogenous signaling pathway that regulates the pro-neural transcription factor Neurogenin 2, is activated by neurotrophins and is critical for adult neurogenesis. We discuss studies demonstrating that specific deletion of ERK5 in the adult neurogenic regions impairs several forms of hippocampus-dependent memory formation in mice. These include contextual fear memory extinction, the establishment and maintenance of remote contextual fear memory, and several other challenging forms of hippocampus-dependent memory formation including 48h memory for novel object recognition, contextual fear memory established by a weak foot shock, pattern separation, and reversal of spatial learning and memory. We also briefly discuss current evidence that increasing adult neurogenesis, by small molecules or genetic manipulation, improves memory formation and long-term memory.


Subject(s)
Dentate Gyrus/physiology , Extinction, Psychological/physiology , Memory/physiology , Mitogen-Activated Protein Kinase 7/metabolism , Neurogenesis , Animals , Dentate Gyrus/cytology , Dentate Gyrus/enzymology , Fear/physiology , Mice , Mice, Knockout , Neurogenesis/genetics
16.
Neurochem Res ; 38(12): 2559-69, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24150751

ABSTRACT

Cyclooxygenase-2 (COX-2) function has been implicated in a number of physiological processes, including inflammatory responses, synaptic transmission, and synaptic plasticity in the brain. However, the specific role of COX-2 in exercise-induced neurogenesis is still debatable. Here, we assessed the role of COX-2 in exercise-induced plasticity by comparing COX-2 knockout mice to wild-type control littermates. We investigated the number of neural stem cells, and the degree of cell proliferation and neuronal differentiation in COX-2 knockout and its wild-type mice that either exercised or remained inactive. Wild-type and COX-2 knockout mice were put on a treadmill and were either sedentary or were forced to run 1 h/day for five consecutive days at a pace of 10-12 m/min for 5 weeks. Loss of COX-2 expression in the knockout mice was confirmed with two measures: (1) COX immunolabeling in the hippocampus, and (2) the identification of abnormal kidney development using hematoxylin and eosin staining, including subcapsular glomerular hypoplasia and hypertrophy of the deeper cortical glomeruli. Compared to wild-type mice, COX-2 knockout mice exhibited a significant reduction in the neural stem cells (nestin-positive cells), cell proliferation (Ki67-positive cells), and neuroblast differentiation (doublecortin-positive cells). In contrast, exercise significantly increased the neural stem cells, cell proliferation, and neuroblast differentiation in both the wild-type and COX-2 knockout mice although the NeuN-immunoreactive neurons were similar in all groups. Expression of phosphorylated cAMP-response element binding protein was decreased in knockout mice. Exercise increased its expression in the subgranular zone of the dentate gyrus in both wild-type and knockout mice. These results suggest that the COX-2 pathway is one of important factors on neural stem cells, cell proliferation and neuroblast differentiation in sedentary mice. The ability of exercise to increase these types of neural plasticity, regardless of COX-2 signaling, suggests that the effects of exercise on neural stem cells, cell proliferation, and neuroblast differentiation are induced via a pathway that is independent of COX-2.


Subject(s)
Cell Differentiation , Cell Proliferation , Cyclooxygenase 2/genetics , Dentate Gyrus/cytology , Neural Stem Cells/cytology , Physical Conditioning, Animal , Walking , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Dentate Gyrus/enzymology , Male , Mice , Mice, Knockout
17.
Neurochem Res ; 38(9): 1980-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23836293

ABSTRACT

Apripiprazole (APZ) is well known as an atypical antipsychotic and antidepressant. In the present study, we investigated effects of APZ on cell proliferation and neuronal differentiation in the dentate gyrus (DG) of the adolescent mouse using BruU, Ki-67 and doublecortin (DCX) immunohistochemistry. BruU, Ki-67 and DCX-positive (+) cells were easily detected in the subgranular zone of the DG in the vehicle- and APZ-treated group. We found that in the 8 mg/kg APZ-treated group numbers of Ki-67(+), DCX(+) and BrdU(+)/DCX(+) cells were significantly increased compared with those in the vehicle-treated group. We also found that maturation and complexity of DCX(+) dendrites in the 8 mg/kg APZ-treated group was well improved compared with those in the vehicle-treated group. In addition, markedly decreased lipid peroxidation and increased superoxide dismutase 2 (SOD2) level were observed in the DG of the 8 mg/kg APZ-treated group. Our present findings indicate that APZ can enhance cell proliferation and neuroblast differentiation, particularly maturation and complexity of neuroblast dendrites, in the DG via decreasing lipid peroxidation and increasing SOD2 level.


Subject(s)
Antipsychotic Agents/pharmacology , Dentate Gyrus/drug effects , Neurons/drug effects , Piperazines/pharmacology , Quinolones/pharmacology , Superoxide Dismutase/metabolism , Animals , Aripiprazole , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dentate Gyrus/cytology , Dentate Gyrus/enzymology , Doublecortin Protein , Male , Mice , Mice, Inbred ICR
18.
Phytother Res ; 27(9): 1293-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23109250

ABSTRACT

Panax ginseng C.A. Meyer has been used in traditional herb prescriptions for thousands of years. A heat-processing method has been used to increase the efficacy of ginseng, yielding what is known as red ginseng. In addition, recently, a slightly modified heat-processing method was applied to ginseng, to obtain a new type of processed ginseng with increased biological activity; this new form of ginseng is referred to as Sun ginseng (SG). The aim of this study was to investigate the effect of SG on memory enhancement and neurogenesis in the hippocampal dentate gyrus (DG) region. The subchronic administration of SG (for 14 days) significantly increased the latency time in the passive avoidance task relative to the administration of the vehicle control (P < 0.05). Western blotting revealed that the levels of phosphorylated extracellular signal-regulated kinase (pERK) and phosphorylated protein kinase B (pAkt) were significantly increased in hippocampal tissue after 14 days of SG administration (P < 0.05). Doublecortin and 5-bromo-2-deoxyuridine immunostaining revealed that SG significantly enhanced the neuronal cell proliferation and the survival of immature neurons in the subgranular zone of the hippocampal DG region. These results suggest that SG has memory-enhancing activities and that these effects are mediated, in part, by the increase in the levels of pERK and pAkt and by the increases in cell proliferation and cell survival.


Subject(s)
Dentate Gyrus/drug effects , Memory/drug effects , Neurogenesis/drug effects , Panax/chemistry , Plant Extracts/pharmacology , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Dentate Gyrus/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Mice , Mice, Inbred ICR , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
19.
J Neurosci ; 31(32): 11472-83, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21832177

ABSTRACT

The dentate gyrus (DG) is one of two areas in the mature brain where stem cells reside to continuously produce new neurons throughout adulthood. While much research has focused on the DG for its roles in adult neurogenesis, little is known regarding how this key region of the brain initially develops to form its distinct architecture. We show here that the murine EphB2 receptor tyrosine kinase is critical for embryonic/postnatal development of a specific region of the DG known as the lateral suprapyramidal blade (LSB). Intracellular truncation and point mutants demonstrate that EphB2 catalytic activity is essential for LSB formation. This is consistent with expression of EphB2 in nestin-positive neural progenitor cells that migrate medially from the lateral ventricle dentate notch neuroepithelium to populate the tertiary matrix and form the DG near the midline of the brain. Animals lacking ephrin-B1 recapitulate loss of the receptor and show that this molecule acts as the ligand to stimulate EphB2 forward signaling and direct migration of the neural progenitors into the dorsal compartment of the tertiary matrix and form the LSB. Immunoreactivity against the extracellular matrix protein Reelin in a region directly above the developing LSB is dramatically reduced when EphB2 forward signaling is disrupted. Together, these results indicate ephrin-B1 interacting with EphB2 controls the migration of dentate progenitor cells into the dorsal half of the developing DG, perhaps in part by affecting Reelin expression in a key compartment directly above the LSB.


Subject(s)
Cell Movement/physiology , Dentate Gyrus/enzymology , Ephrin-B1/physiology , Neurons/enzymology , Receptor, EphB2/physiology , Signal Transduction/physiology , Stem Cells/enzymology , Animals , Dentate Gyrus/embryology , Dentate Gyrus/growth & development , Female , Ligands , Mice , Mice, Knockout , Mice, Mutant Strains , Neurons/cytology , Pregnancy , Reelin Protein , Stem Cells/cytology
20.
J Neurochem ; 123(5): 676-88, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23020821

ABSTRACT

The neurogenic niche within the subgranular zone (SGZ) of the dentate gyrus is a source of new neurons throughout life. Interestingly, SGZ proliferative capacity is regulated by both physiological and pathophysiological conditions. One outstanding question involves the molecular mechanisms that regulate both basal and inducible adult neurogenesis. Here, we examined the role of the MAPK-regulated kinases, mitogen- and stress-activated kinase (MSK)1 and MSK2. as regulators of dentate gyrus SGZ progenitor cell proliferation and neurogenesis. Under basal conditions, MSK1/2 null mice exhibited significantly reduced progenitor cell proliferation capacity and a corollary reduction in the number of doublecortin (DCX)-positive immature neurons. Strikingly, seizure-induced progenitor proliferation was totally blocked in MSK1/2 null mice. This blunting of cell proliferation in MSK1/2 null mice was partially reversed by forskolin infusion, indicating that the inducible proliferative capacity of the progenitor cell population was intact. Furthermore, in MSK1/2 null mice, DCX-positive immature neurons exhibited reduced neurite arborization. Together, these data reveal a critical role for MSK1/2 as regulators of both basal and activity-dependent progenitor cell proliferation and morphological maturation in the SGZ.


Subject(s)
Adult Stem Cells/enzymology , Cell Proliferation , Neural Stem Cells/enzymology , Neurogenesis/physiology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Adult Stem Cells/cytology , Animals , Dentate Gyrus/cytology , Dentate Gyrus/enzymology , Doublecortin Protein , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Knockout , Ribosomal Protein S6 Kinases, 90-kDa/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL