Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Country/Region as subject
Publication year range
1.
FASEB J ; 34(2): 3267-3288, 2020 02.
Article in English | MEDLINE | ID: mdl-31908036

ABSTRACT

TIAM2S, the short form of human T-cell lymphoma invasion and metastasis 2, can have oncogenic effects when aberrantly expressed in the liver or lungs. However, it is also abundant in healthy, non-neoplastic brain tissue, in which its primary function is still unknown. Here, we examined the neurobiological and behavioral significance of human TIAM2S using the human brain protein panels, a human NT2/D1-derived neuronal cell line model (NT2/N), and transgenic mice that overexpress human TIAM2S (TIAM2S-TG). Our data reveal that TIAM2S exists primarily in neurons of the restricted brain areas around the limbic system and in well-differentiated NT2/N cells. Functional studies revealed that TIAM2S has no guanine nucleotide exchange factor (GEF) activity and is mainly located in the nucleus. Furthermore, whole-transcriptome and enrichment analysis with total RNA sequencing revealed that TIAM2S-knockdown (TIAM2S-KD) was strongly associated with the cellular processes of the brain structural development and differentiation, serotonin-related signaling, and the diseases markers representing neurobehavioral developmental disorders. Moreover, TIAM2S-KD cells display decreased neurite outgrowth and reduced serotonin levels. Moreover, TIAM2S overexpressing TG mice show increased number and length of serotonergic fibers at early postnatal stage, results in higher serotonin levels at both the serum and brain regions, and higher neuroplasticity and hyperlocomotion in latter adulthood. Taken together, our results illustrate the non-oncogenic functions of human TIAM2S and demonstrate that TIAM2S is a novel regulator of serotonin level, brain neuroplasticity, and locomotion behavior.


Subject(s)
Brain/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Locomotion , Serotonin/metabolism , Animals , Brain/growth & development , Brain/physiology , Cell Line, Tumor , Cells, Cultured , Guanine Nucleotide Exchange Factors/genetics , Humans , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neuronal Outgrowth , Neuronal Plasticity
2.
Int J Mol Sci ; 22(19)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34638842

ABSTRACT

Despite neurosurgery following radiation and chemotherapy, residual glioblastoma (GBM) cells develop therapeutic resistance (TR) leading to recurrence. The GBM heterogeneity confers TR. Therefore, an effective strategy must target cancer stem cells (CSCs) and other malignant cancer cells. TGF-ß and mesenchymal transition are the indicators for poor prognoses. The activity of aldehyde dehydrogenases (ALDHs) is a functional CSC marker. However, the interplay between TGF-ß and ALDHs remains unclear. We developed radiation-resistant and radiation-temozolomide-resistant GBM models to investigate the underlying mechanisms conferring TR. Galunisertib is a drug targeting TGF-ß receptors. Disulfiram (DSF) is an anti-alcoholism drug which functions by inhibiting ALDHs. The anti-tumor effects of combining DSF and Galunisertib were evaluated by in vitro cell grow, wound healing, Transwell assays, and in vivo orthotopic GBM model. Mesenchymal-like phenotype was facilitated by TGF-ß in TR GBM. Additionally, TR activated ALDHs. DSF inhibited TR-induced cell migration and tumor sphere formation. However, DSF did not affect the tumor growth in vivo. Spectacularly, DSF sensitized TR GBM to Galunisertib both in vitro and in vivo. ALDH activity positively correlated with TGF-ß-induced mesenchymal properties in TR GBM. CSCs and mesenchymal-like GBM cells targeted together by combining DSF and Galunisertib may be a good therapeutic strategy for recurrent GBM patients.


Subject(s)
Disulfiram/pharmacology , Drug Resistance, Neoplasm/drug effects , Glioblastoma/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Radiation Tolerance/drug effects , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Temozolomide/pharmacology , Animals , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism
3.
J Biomed Sci ; 25(1): 3, 2018 Jan 11.
Article in English | MEDLINE | ID: mdl-29325565

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive memory and neuronal loss culminating in cognitive impairment that not only affects a person's living ability but also becomes a society's as well as a family's economic burden. AD is the most common form of dementia in older persons. It is expected that the number of people with AD dementia will increase dramatically in the next 30 years, projecting to 75 million in 2030 and 131.5 million in 2050 worldwide. So far, no sufficient evidence is available to support that any medicine is able to prevent or reverse the progression of the disease. Early studies have shown that social environment, particularly social relationships, can affect one's behavior and mental health. A study analyzing the correlation between loneliness and risk of developing AD revealed that lonely persons had higher risk of AD compared with persons who were not lonely. On the other hand, it has been reported that we can prevent cognitive decline and delay the onset of AD if we keep mentally active and frequently participate in social activities. In this review, we focus on the impact of social behaviors on the progression of cognitive deficit in animal models of AD with a particular emphasis on a mechanistic scheme that explains how social isolation exacerbates cognitive impairment and how social interaction with conspecifics rescues AD patients' memory deficit.


Subject(s)
Alzheimer Disease/prevention & control , Cognitive Dysfunction/prevention & control , Disease Progression , Interpersonal Relations , Social Isolation , Alzheimer Disease/psychology , Animals , Cognitive Dysfunction/psychology , Disease Models, Animal , Humans , Mice , Primates , Rats
4.
Neurobiol Learn Mem ; 141: 1-8, 2017 May.
Article in English | MEDLINE | ID: mdl-28274822

ABSTRACT

Co-housing with a company exerts profound effects on memory decline in animal model of Alzheimer's disease (AD). Recently, we found that APP/PS1 mice of 9-month-old improved their memories after co-housing with wide-type mice for 3months by increasing hippocampal brain-derived neurotrophic factor (BDNF) expression. However, the mechanism of how co-housing could induce BDNF expression remains elusive. Here we examined epigenetic changes in the mouse hippocampus that accompanied the co-housing-induced memory improvement. We found that the level of histone deacetylase 2 (HDAC2), but not that of HDAC1, was significantly lower in the memory improved mice than in the control and memory un-improved APP/PS1 mice after co-housing. Knockdown of Hdac2 resulted in a higher freezing response after co-housing. Conversely, over-expression of HDAC2 blocked co-housing-induced memory improvement. The level of Bdnf exon IV mRNA increased significantly after knockdown of Hdac2. ChIP assay revealed a decreased occupancy of HDAC2 in the promoter region of Bdnf exon IV of memory improved mice but not memory un-improved and control APP/PS1 mice. Consistently, the acetylation of histone 3 on Lys 9 (H3K9) and histone 4 on Lys12 (H4K12) increased significantly in the promoter region of Bdnf exon IV. These results suggest HDAC2 expression is reduced after co-housing resulting in a decreased occupancy of HDAC2 and increased histone H3K9 and H4K12 acetylation in the promoter region of Bdnf exon IV, leading to increased BDNF expression in the hippocampus that improves memory.


Subject(s)
Alzheimer Disease/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Epigenesis, Genetic , Hippocampus/metabolism , Housing, Animal , Memory Disorders/metabolism , Alzheimer Disease/psychology , Animals , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Male , Memory Disorders/psychology , Mice
5.
BMC Cancer ; 17(1): 905, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29284440

ABSTRACT

BACKGROUND: Glioma stem cells (GSCs) contribute to tumor recurrence and drug resistance. This study characterizes the tumorigenesis of CD133+ cells and their sensitivity to pharmacological inhibition. METHODS: GSCs from human U87 and rat C6 glioblastoma cell lines were isolated via magnetic cell sorting using CD133 as a cancer stem cell marker. Cell proliferation was determined using the WST-1 assay. An intracranial mouse model and bioluminescence imaging were used to assess the effects of drugs on tumor growth in vivo. RESULTS: CD133+ cells expressed stem cell markers and exhibited self-renewal and enhanced tumor formation. Minocycline (Mino) was more effective in reducing the survival rate of CD133+ cells, whereas CD133- cells were more sensitive to inhibition by the signal transducer and activator of transcription 3 (STAT3) inhibitor. Inhibition of STAT3 decreased the expression of CD133+ stem cell markers. The combination of Mino and STAT3 inhibitor synergistically reduced the cell viability of glioma cells. Furthermore, this combination synergistically suppressed tumor growth in nude mice. CONCLUSION: The results suggest that concurrent targeting of different subpopulations of glioblastoma cells may be an effective therapeutic strategy for patients with malignant glioma.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Drug Synergism , Glioblastoma/drug therapy , Minocycline/pharmacology , Neoplastic Stem Cells/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , AC133 Antigen/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Drug Combinations , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Rats , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Learn Mem ; 23(9): 486-93, 2016 09.
Article in English | MEDLINE | ID: mdl-27531839

ABSTRACT

Destabilization refers to a memory that becomes unstable when reactivated and is susceptible to disruption by amnestic agents. Here we delineated the cellular mechanism underlying the destabilization of drug memory. Mice were conditioned with methamphetamine (MeAM) for 3 d, and drug memory was assessed with a conditioned place preference (CPP) protocol. Anisomycin (ANI) was administered 60 min after the CPP retrieval to disrupt reconsolidation. We found that destabilization of MeAM CPP after the application of ANI was blocked by the N-methyl-d-aspartate receptor (NMDAR) antagonist MK-801 and the NR2B antagonist ifenprodil (IFN) but not by the NR2A antagonist NVP-AAM077 (NVP). In addition, decrease in the phosphorylation of GluR1 at Serine845 (p-GluR1-Ser845), decrease in spine density, and a reduction in the AMPAR/NMDAR ratio in the basolateral amygdala (BLA) were reversed after the MK-801 treatment. The effect of ANI on destabilization was prevented by the protein phosphatase 2B (calcineurin, CaN) inhibitors cyclosporine A (CsA) and FK-506 and the protein phosphatase 1 (PP1) inhibitors calyculin A (CA) and okadaic acid (OA). These results suggest that memory destabilization involves the activation of NR2B-containing NMDARs, which in turn allows the influx of Ca(2+) Increased intracellular Ca(2+) stimulates CaN, leading to the dephosphorylation and inactivation of inhibitor 1 and the activation of PP1. PP1 then dephosphorylates p-GluR1-Ser845 to elicit AMPA receptor (AMPAR) endocytosis and destabilization of the drug memory.


Subject(s)
Amygdala/enzymology , Memory Consolidation/physiology , Methamphetamine/administration & dosage , Phosphoprotein Phosphatases/physiology , Amygdala/drug effects , Animals , Anisomycin/administration & dosage , Calcium Signaling/drug effects , Conditioning, Classical , Dendritic Spines/drug effects , Dendritic Spines/physiology , Dizocilpine Maleate/administration & dosage , Male , Memory Consolidation/drug effects , Mental Recall/drug effects , Mental Recall/physiology , Mice, Inbred C57BL , Phosphoprotein Phosphatases/antagonists & inhibitors , Protein Synthesis Inhibitors/administration & dosage , Quinoxalines/administration & dosage , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology
7.
J Neurochem ; 137(2): 216-25, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26748780

ABSTRACT

Addiction is thought to be a memory process between perception and environmental cues and addicted patients often relapse when they come into contact with the drug-related context once again. Here, we used a conditioned place preference protocol to seek a more effective extinction methodology of methamphetamine (METH) memory and delineate its underlying mechanism. Conditioning METH for 3 days in mice markedly increased the time spent in the METH-paired compartment. Then the mice were conditioned with saline for 6 days, from day 6 to day 11, a procedure termed extinction training. However, METH memory returned after a priming injection of METH. We prolonged extinction duration from 6 to 10 days and found that this extensive extinction (EE) training prevented priming effect. At the molecular level, we discovered that prolonged extinction training reversed the METH-conditioned place preference-induced increase in surface expression of GluA2 and alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA)/NMDA ratio in the basolateral amygdala. In addition, we found that extinction with metabotropic glutamate receptor 5 (mGluR5) activation had similar results to EE: reduced relapse after extinction, decreased synaptic AMPA receptors AMPARs and the AMPA/NMDA ratio. On the contrary, EE with mGluR5 inhibition suppressed the results of EE. These data indicate that EE training-elicited inhibition of METH-primed reinstatement is mediated by the mGluR5. Conditioning mice with methamphetamine place preference (METH CPP) increases surface expression of AMPA receptors (AMPARs) in the basolateral amygdala. We found prolongation of extinction duration from 6 to 10 days prevented priming effect. At the molecular level, we discovered that extensive extinction (EE) reversed the METH CPP-induced increase in surface expression of GluA2 and AMPA/NMDA ratio. In addition, we found that extinction with the metabotropic glutamate receptor 5 (mGluR5) activation had similar results to EE: reduced relapse after extinction, decreased synaptic AMPARs and the AMPA/NMDA ratio. On the contrary, EE with mGluR5 inhibition suppressed the results of EE. These data indicate that EE training-elicited inhibition of METH-primed reinstatement is mediated by mGluR5 (PAM: positive allosteric modulator).


Subject(s)
Central Nervous System Stimulants/pharmacology , Conditioning, Operant/drug effects , Extinction, Psychological/drug effects , Methamphetamine/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism , Action Potentials/drug effects , Animals , Benzamides/pharmacology , Bicuculline/pharmacology , Biotinylation , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Exploratory Behavior/drug effects , GABA-A Receptor Antagonists/pharmacology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Pyrazoles/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
8.
J Biomed Sci ; 23: 10, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26786523

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is the most lethal primary brain tumors which remains difficult to cure despite advances in surgery, radiotherapy and chemotherapy. Therefore, the development of new drug is urgently needed. α-carboline derivatives were usually isolated from marine animals such as Britannia marine tunicate Dendrodoa grossularia and Indonesian ascidian Polycarpa aurata. In this study, we have synthesized several α-carboline compounds and examined their anti-glioma activities. RESULTS: We report that among α-carboline derivatives TJY-16 (6-acetyl-9-(3,4,5-trimethoxybenzyl)-9H-pyrido[2,3-b] indole) is the most potent α-carboline analog to induce glioma cell death with IC50 value of around 50 nM. TJY-16 decreased cell viability of glioma cells in a concentration- and time-dependent manner. Trypan blue exclusion assay showed that the reduction of cell viability was due to both cell growth inhibition and cell death. Flow cytometric analysis showed that TJY-16 induced G2/M cell cycle arrest followed by induction of sub-G1 phase. Hoechst staining detected the apoptotic features such as nuclear shrinkage and DNA condensation. Western blot analysis showed the increased level of cleaved caspase-3. The activation of caspase-8 and depolarization of mitochondrial membrane potential (ΔΨm) indicated that both extrinsic and intrinsic apoptotic pathways were involved in TJY-16-induced apoptosis. TJY-16 effectively inhibited tumor growth and induced caspase-3 activation in the xenograft tumor model of U87 glioma cells. CONCLUSIONS: Our results suggest that TJY-16 may kill glioma cells by inducing G2/M cell cycle arrest followed by apoptosis. Thus, TJY-16 is a promising agent for the treatment of malignant gliomas.


Subject(s)
Antineoplastic Agents , Apoptosis/drug effects , DNA, Neoplasm/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Glioma , M Phase Cell Cycle Checkpoints/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Glioma/drug therapy , Glioma/metabolism , Glioma/pathology , Humans , Urochordata/chemistry
9.
J Neurosci ; 34(49): 16207-19, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25471562

ABSTRACT

It has been recognized that the risk of cognitive decline during aging can be reduced if one maintains strong social connections, yet the neural events underlying this beneficial effect have not been rigorously studied. Here, we show that amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic (APP/PS1) mice demonstrate improvement in memory after they are cohoused with wild-type mice. The improvement was associated with increased protein and mRNA levels of BDNF in the hippocampus. Concomitantly, the number of BrdU(+)/NeuN(+) cells in the hippocampal dentate gyrus was significantly elevated after cohousing. Methylazoxymethanol acetate, a cell proliferation blocker, markedly reduced BrdU(+) and BrdU/NeuN(+) cells and abolished the effect of social interaction. Selective ablation of mitotic neurons using diphtheria toxin (DT) and a retrovirus vector encoding DT receptor abolished the beneficial effect of cohousing. Knockdown of BDNF by shRNA transfection blocked, whereas overexpression of BDNF mimicked the memory-improving effect. A tropomyosin-related kinase B agonist, 7,8-dihydroxyflavone, occluded the effect of social interaction. These results demonstrate that increased BDNF expression and neurogenesis in the hippocampus after cohousing underlie the reversal of memory deficit in APP/PS1 mice.


Subject(s)
Alzheimer Disease/therapy , Brain-Derived Neurotrophic Factor/physiology , Hippocampus/metabolism , Memory Disorders/therapy , Neurogenesis/physiology , Social Behavior , Alzheimer Disease/complications , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Dentate Gyrus/physiology , Diphtheria Toxin/pharmacology , Disease Models, Animal , Flavones/pharmacology , Gene Knockdown Techniques , Hippocampus/growth & development , Housing, Animal , Male , Memory Disorders/complications , Methylazoxymethanol Acetate/pharmacology , Mice , Mice, Transgenic , Neurogenesis/drug effects , Presenilin-1/genetics
10.
Hippocampus ; 25(4): 474-85, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25348768

ABSTRACT

Epidemiological studies have shown that early life adverse events have long-term effects on the susceptibility to subsequent stress exposure in adolescence, but the precise mechanism is unclear. In the present study, mice on postnatal day 21-28 were randomly assigned to either a group or isolated cages for 8 weeks. The socially isolated (SI) mice exhibited a higher level of spontaneous locomotor activity, a longer duration of immobility in the forced swimming test (FST), significantly less prepulse inhibition (PPI) and an increase in aggressive (but not attack) behavior. However, acute stress markedly exacerbated the attack counts of the SI mice but did not affect the group housing (GH) mice. SI mice exhibited higher synaptosomal NR2A and NR2B levels in the hippocampus as compared to the GH mice. Whole-cell patch clamp recordings of CA1 neurons in hippocampal slices showed that the SI mice exhibited a higher input-output relationship of NMDAR-EPSCs as compared to the GH mice. Application of the NR2B -specific antagonist ifenprodil produced a greater attenuating effect on NMDAR-EPSCs in slices from the SI mice. NMDAR EPSCs recorded from the SI mice had a slower deactivation kinetic. MK-801, CPP and ifenprodil, the NMDA antagonists, reversed acute stress-induced exaggeration of aggressive and depressive behaviors. Furthermore, acute stress-induced exacerbation of attack behavior in the SI mice was abolished after the knockdown of NR2B expression. These results suggest that social isolation-induced increased expression of NMDA receptors in the hippocampus involves stress exacerbation of aggressive behaviors. Amelioration of aggressive behaviors by NMDA antagonists may open a new avenue for the treatment of psychopathologies that involve outbursts of emotional aggression in neglected children.


Subject(s)
Affective Symptoms/etiology , Affective Symptoms/pathology , Hippocampus/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Social Isolation/psychology , Aggression/physiology , Animals , Animals, Newborn , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Exploratory Behavior/physiology , Gene Expression Regulation/physiology , Hippocampus/cytology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Piperidines/pharmacology , Prepulse Inhibition/drug effects , Prepulse Inhibition/physiology , Random Allocation , Reflex, Startle/physiology , Swimming/psychology , Synaptosomes/drug effects , Synaptosomes/metabolism
11.
Eur J Neurosci ; 39(1): 83-96, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24206109

ABSTRACT

We used the oxygen and glucose deprivation (OGD) method in cultured astrocytes as an in vitro ischemic model. We investigated whether activation of group-II metabotropic glutamate receptors (mGluR2/3) can reverse OGD-induced impairment in astrocytic glutamate/aspartate transporter (GLAST) expression and elucidated the signaling pathways involving the GLAST expression. Cultured astrocytes exposed to OGD for 6 h resulted in significant reductions in the GLAST expression and extracellular glutamate clearance. These reductions were effectively restored by mGluR2/3 activation with mGluR2/3 agonists, LY379268 or DCG-IV, after the 6 h OGD insult. These mGluR2/3-mediated restorative effects were inhibited by selective mGluR2/3 antagonists LY341459 or EGLU. The mGluR2/3 activation also induced activations of signaling pathways including extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K) and nuclear transcription factor-κB (NFκB). These activations were prevented by blocking mGluR2/3 with LY341459, an mGluR2/3 antagonist. Furthermore, blocking ERK, PI3K and NFκB signaling pathways with U0126, LY294002 and pyrrolidine dithiocarbamate, respectively, significantly inhibited the mGluR2/3-mediated restorative effects. These results suggest that application of mGluR2/3 agonists after OGD insult can effectively reverse the OGD-reduced expression of GLAST proteins and restore clearance of extracellular glutamate by serially activating ERK/PI3K/NFκB signaling pathways in cultured astrocytes.


Subject(s)
Astrocytes/metabolism , Glucose/deficiency , Glutamic Acid/metabolism , MAP Kinase Signaling System , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Astrocytes/drug effects , Cell Hypoxia , Cells, Cultured , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Transporter 1/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism
12.
Int J Neuropsychopharmacol ; 18(3)2014 Dec 13.
Article in English | MEDLINE | ID: mdl-25522410

ABSTRACT

BACKGROUND: It is known that neurogenesis occurs throughout the life mostly in the subgranular zone of the hippocampus and the subventricular zone of the lateral ventricle. We investigated whether neurogenesis occurred in the amygdala and its function in fear memory formation. METHODS: For detection of newborn neurons, mice were injected intraperitoneally with 5-bromo-2'-deoxyuridine (BrdU) 2h before receiving 15 tone-footshock pairings, and newborn neurons were analyzed 14 and 42 days after training. To determine the relationship between neurogenesis and memory formation, mice were given a proliferation inhibitor methylazoxymethanol (MAM) or a DNA synthesis inhibitor cytosine arabinoside (Ara-C). To test whether sonic hedgehog (Shh) signaling was required for neurogenesis, Shh-small hairpin-interfering RNA (shRNA) was inserted into a retroviral vector (Retro-Shh-shRNA). RESULTS: The number of BrdU(+)/Neuronal nuclei (NeuN)(+) cells was significantly higher in the conditioned mice, suggesting that association of tone with footshock induced neurogenesis. MAM and Ara-C markedly reduced neurogenesis and impaired fear memory formation. Shh, its receptor patched 1 (Ptc1), and transcription factor Gli1 protein levels increased at 1 day and returned to baseline at 7 days after fear conditioning. Retro-Shh-shRNA, which knocked down Shh specifically in the mitotic neurons, reduced the number of BrdU(+)/NeuN(+) cells and decreased freezing responses. CONCLUSIONS: These results suggest that fear learning induces Shh signaling activation in the amygdala, which promotes neurogenesis and fear memory formation.


Subject(s)
Amygdala/metabolism , Conditioning, Classical/physiology , Hedgehog Proteins/metabolism , Memory, Long-Term/physiology , Neurogenesis/physiology , Signal Transduction/physiology , Animals , Bromodeoxyuridine , Cytarabine/pharmacology , Electroshock , Exploratory Behavior , Fear/psychology , Immunosuppressive Agents/pharmacology , Long-Term Potentiation , Male , Methylazoxymethanol Acetate/analogs & derivatives , Methylazoxymethanol Acetate/pharmacology , Mice , Mice, Inbred C57BL , RNA, Small Interfering/pharmacology
13.
Horm Behav ; 66(3): 498-508, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25066484

ABSTRACT

Women are thought to form fear memory more robust than men do and testosterone is suspected to play a role in determining such a sex difference. Mouse cued fear freezing was used to study the sex-related susceptibility and the role of testosterone in fear memory in humans. A 75-dB tone was found to provoke weak freezing, while 0.15-mA and 0.20-mA footshock caused strong freezing responses. No sex differences were noticed in the tone- or footshock-induced (naïve fear) freezing. Following the conditionings, female mice exhibited greater tone (cued fear)-induced freezing than did male mice. Nonetheless, female mice demonstrated indistinctive cued fear freezing across the estrous phases and ovariectomy did not affect such freezing in female mice. Orchidectomy enhanced the cued fear freezing in male mice. Systemic testosterone administrations and an intra-lateral nucleus of amygdala (LA) testosterone infusion diminished the cued fear freezing in orchidectomized male mice, while pretreatment with flutamide (Flu) eradicated these effects. Long-term potentiation (LTP) magnitude in LA has been known to correlate with the strength of the cued fear conditioning. We found that LA LTP magnitude was indeed greater in female than male mice. Orchidectomy enhanced LTP magnitude in males' LA, while ovariectomy decreased LTP magnitude in females' LA. Testosterone decreased LTP magnitude in orchidectomized males' LA and estradiol enhanced LTP magnitude in ovariectomized females' LA. Finally, male mice had lower LA GluR1 expression than female mice and orchidectomy enhanced the GluR1 expression in male mice. These findings, taken together, suggest that testosterone plays a critical role in rendering the sex differences in the cued fear freezing and LA LTP. Testosterone is negatively associated with LA LTP and the cued fear memory in male mice. However, ovarian hormones and LA LTP are loosely associated with the cued fear memory in female mice.


Subject(s)
Amygdala/physiology , Fear/psychology , Long-Term Potentiation/physiology , Memory/physiology , Testosterone/physiology , Animals , Conditioning, Psychological/drug effects , Cues , Fear/drug effects , Fear/physiology , Female , Freezing Reaction, Cataleptic/drug effects , Long-Term Potentiation/drug effects , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Sex Characteristics , Testosterone/pharmacology
14.
Learn Mem ; 20(6): 328-35, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23685808

ABSTRACT

Leptin, a 167 amino acid peptide, is synthesized predominantly in the adipose tissues and plays a key role in the regulation of food intake and body weight. Recent studies indicate that leptin receptor is expressed with high levels in many brain regions that may regulate synaptic plasticity. Here we show that deprivation of rapid eye movement (REMD) sleep resulted in impairment of both cue and contextual fear memory. In parallel, surface expression of GluR1 was reduced in the amygdala. Intraperitoneal injection of leptin to the REMD mice rescued memory impairment and reversed surface GluR1 reduction. Using whole-cell recording to evaluate the synaptic function of the thalamus-lateral amygdala (LA) pathway, we found a decrease in frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs) concomitant with reduced AMPA/NMDA ratios in the REMD mice. By contrast, paired-pulse facilitation (PPF) was increased. The effects of REMD on mEPSCs and AMPA/NMDA ratio could be reversed by leptin treatment, whereas on PPR it could not. Phosphatase and tensin homolog (PTEN), a dual protein/lipid phosphatase, down-regulates the effect of the PI-3 kinase pathway. Fear conditioning increased whereas REMD led to a decrease in the phosphorylated states of PTEN, Akt, and glycogen synthase kinase-3ß (GSK3ß), and the effects of REMD were reversed by leptin. These results suggest that both pre- and postsynaptic functions of the thalamus-LA pathway were altered by fear conditioning and REMD in opposite directions. Leptin treatment reversed REMD-induced memory deficits primarily by a postsynaptic action by restoring surface expression of GluR1 without affecting PPR.


Subject(s)
Brain/metabolism , Leptin/metabolism , Memory/physiology , Sleep Deprivation/complications , Sleep Deprivation/metabolism , Animals , Blotting, Western , Conditioning, Classical , Excitatory Postsynaptic Potentials/physiology , Male , Mice , Mice, Inbred C57BL , Neural Pathways/metabolism , Neuronal Plasticity/physiology , Patch-Clamp Techniques
15.
Neurobiol Learn Mem ; 103: 72-81, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23603364

ABSTRACT

Repetitive drug taking induces neural long-lasting changes and results in compulsive drug-seeking behavior which may arise from enduring drug memory that impairs cognitive control of motivated behavior. Thus, disrupting these memories could reduce drug seeking. Here, we used a conditioned place preference (CPP) procedure in mice to examine the role of AMPA receptor endocytosis in the basolateral amygdala (BLA) in the disrupted reconsolidation of Methamphetamine (MeAM) memory. Conditioning MeAM (2mg/kg, i.p.) for 3 days in mice markedly increased the time spent in the MeAM-paired compartment tested 24 h after the last injection (CPP test), indicating that MeAM induced a significant rewarding effect. Mice then received anisomycin or vehicle within 1h after CPP test and CPP was re-assessed 24h after CPP test. Mice injected with vehicle exhibited CPP for the previously MeAM-paired chamber whereas mice injected with anisomycin did not. Anisomycin had no effect on the CPP when CPP test was omitted. In addition, anisomycin treatment prevented MeAM priming-induced reinstatement of CPP suggesting the disruption of MeAM memory reconsolidation. MeAM CPP increased surface expression of GluR1 and GluR2 subunits of AMPA receptor in the BLA. Bilateral injection of Tat-GluR23Y, a synthetic peptide that blocked AMPA receptor endocytosis, prevented disruption of MeAM memory reconsolidation. These results suggest that AMPA receptor endocytosis in the BLA is critical for the anisomycin-mediated disruption of reconsolidation of MeAM reward memory.


Subject(s)
Amygdala/metabolism , Association Learning/drug effects , Dopamine Uptake Inhibitors/pharmacology , Endocytosis/physiology , Memory/drug effects , Methamphetamine/pharmacology , Receptors, AMPA/metabolism , Amygdala/drug effects , Animals , Anisomycin/pharmacology , Association Learning/physiology , Conditioning, Operant/drug effects , Drug-Seeking Behavior/drug effects , Drug-Seeking Behavior/physiology , Endocytosis/drug effects , Extinction, Psychological/drug effects , Male , Memory/physiology , Mice , Protein Synthesis Inhibitors/pharmacology
16.
Int J Neuropsychopharmacol ; 16(9): 2027-39, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23823694

ABSTRACT

Accumulating evidence suggests that dysfunction of the amygdala is related to abnormal fear processing, anxiety, and social behaviors noted in autistic spectrum disorders (ASDs). In addition, studies have shown that disrupted brain serotonin homeostasis is linked to ASD. With a valproate (VPA)-induced rat ASD model, we investigated the possible role of amygdala serotonin homeostasis in autistic phenotypes and further explored the underlying mechanism. We first discovered that the distribution of tryptophan hydroxylase immunoreactivity in the caudal raphe system was modulated on postnatal day (PD) 28 of the VPA-exposed offspring. Then, we found a significantly higher serotonin transporter availability in the amygdala of the VPA-exposed offspring on PD 56 by using single photon emission computed tomography and computed tomography co-registration following injection of (123)I-labeled 2-((2-(dimethylamino)methyl)phenyl)thio)-5-iodophenylamine((123)I[ADAM]). Furthermore, treatment with 8-hydroxy-2-(di-n-propylamino)tetralin (8-OH-DPAT), a 5-HT1A receptor agonist, increased social interaction and improved fear memory extinction in the VPA-exposed offspring. 8-OH-DPAT treatment also reversed the characteristics of miniature excitatory post-synaptic currents as well as paired pulse facilitation observed in lateral amygdala slices. These results provided further evidence to support the role of the amygdala in characteristic behavioral changes in the rat ASD model. The serotonergic projections that modulate the amygdala function might play a certain role in the development and treatment of behavioral symptoms exhibited in individuals with ASD.


Subject(s)
8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Amygdala/drug effects , Autistic Disorder/drug therapy , Behavior, Animal/drug effects , Receptor, Serotonin, 5-HT1A/drug effects , Serotonin 5-HT1 Receptor Agonists/pharmacology , Social Behavior , Valproic Acid , Amygdala/diagnostic imaging , Amygdala/metabolism , Amygdala/physiopathology , Animals , Autistic Disorder/chemically induced , Autistic Disorder/diagnosis , Autistic Disorder/metabolism , Autistic Disorder/physiopathology , Autistic Disorder/psychology , Cinanserin/analogs & derivatives , Disease Models, Animal , Excitatory Postsynaptic Potentials , Extinction, Psychological/drug effects , Fear/drug effects , Female , Male , Memory/drug effects , Miniature Postsynaptic Potentials , Multimodal Imaging/methods , Pregnancy , Prenatal Exposure Delayed Effects , Radiopharmaceuticals , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Time Factors , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Tryptophan Hydroxylase/metabolism
17.
Nucleic Acids Res ; 39(13): 5412-23, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21441538

ABSTRACT

The exact mechanism underlying increases in Sp1 and the physiological consequences thereafter remains unknown. In rat primary cortical neurons, oxygen-glucose deprivation (OGD) causes an increase in H(2)O(2) as well as Sp1 in early ischaemia but apparently does not change mRNA level or Sp1 stability. We hereby identified a longer 5'-UTR in Sp1 mRNA that contains an internal ribosome entry site (IRES) that regulates rapid and efficient translation of existing mRNAs. By using polysomal fragmentation and bicistronic luciferase assays, we found that H(2)O(2) activates IRES-dependent translation. Thus, H(2)O(2) or tempol, a superoxide dismutase-mimetic, increases Sp1 levels in OGD-treated neurons. Further, early-expressed Sp1 binds to Sp1 promoter to cause a late rise in Sp1 in a feed-forward manner. Short hairpin RNA against Sp1 exacerbates OGD-induced apoptosis in primary neurons. While Sp1 levels increase in the cortex in a rat model of stroke, inhibition of Sp1 binding leads to enhanced apoptosis and cortical injury. These results demonstrate that neurons can use H(2)O(2) as a signalling molecule to quickly induce Sp1 translation through an IRES-dependent translation pathway that, in cooperation with a late rise in Sp1 via feed-forward transcriptional activation, protects neurons against ischaemic damage.


Subject(s)
5' Untranslated Regions , Brain Ischemia/metabolism , Hydrogen Peroxide/pharmacology , Protein Biosynthesis , Sp1 Transcription Factor/genetics , Animals , Glucose/physiology , Humans , Male , Neurons/metabolism , Oxygen/physiology , Rats , Ribosomes/metabolism , Sp1 Transcription Factor/biosynthesis , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Transcriptional Activation
18.
Neuropsychopharmacology ; 48(3): 478-488, 2023 02.
Article in English | MEDLINE | ID: mdl-36109595

ABSTRACT

Drug-associated conditioned cues promote subjects to recall drug reward memory, resulting in drug-seeking and reinstatement. A consolidated memory becomes unstable after recall, such that the amnestic agent can disrupt the memory during the reconsolidation stage, which implicates a potential therapeutic strategy for weakening maladaptive memories. The basolateral amygdala (BLA) involves the association of conditioned cues with reward and aversive valences and projects the information to the nucleus accumbens (NAc) that mediates reward-seeking. However, whether the BLA-NAc projection plays a role in drug-associated memory reactivation and reconsolidation is unknown. We used methamphetamine (MeAM) conditioned place preference (CPP) to investigate the role of BLA-NAc neural projection in the memory reconsolidation. Two weeks before CPP training, we infused adeno-associated virus (AAV) carrying the designer receptor exclusively activated by designer drugs (DREADD) or control constructs. We infused clozapine-N-oxide (CNO) after the recall test to manipulate the neural activity of BLA-NAc projections in mice. We found that after recall, DREADD-mediated inhibition of BLA neurons projecting to the NAc core blunted consolidated MeAM-associated memory. Inhibition of BLA glutamatergic nerve terminals in the NAc core 1 h after recall disrupted consolidated MeAM-associated memory. However, inhibiting this pathway after the time window of reconsolidation failed to affect memory. Furthermore, under the condition without memory retrieval, DREADD-mediated activation of BLA-NAc core projection was required for amnesic agents to disrupt consolidated MeAM-associated memory. Our findings provide evidence that the BLA-NAc pathway activity is involved in the post-retrieval processing of MeAM-associated memory in CPP.


Subject(s)
Basolateral Nuclear Complex , Methamphetamine , Mice , Animals , Methamphetamine/pharmacology , Methamphetamine/metabolism , Amygdala/metabolism , Nucleus Accumbens/metabolism , Memory/physiology
19.
World Neurosurg ; 180: e350-e363, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37757945

ABSTRACT

BACKGROUND: The beneficial effect of pretreatment with statins on traumatic brain injury (TBI)-induced depression and anxiety and its mechanism of action remain unclear. In this study, we combined epidemiological and experimental animal data to clarify this issue. METHODS: We used the Taiwan National Health Insurance database to identify patients who were diagnosed with TBI from 2000 to 2013 and compared patients with and without statin treatment matched by age, sex, and underlying comorbidities in a 1:1 ratio. The risk of developing depression and/or anxiety was compared between patients with and without a statin using Cox proportional hazards regression. We also used a rat model to assess the effect of lovastatin pretreatment on neurobehavioral and neuropathological changes following TBI. RESULTS: The risk of developing depression was lower in the 41,803 patients in the statin cohort than nonstatin cohort (adjusted hazard ratio, 0.91 [95% confidence interval, 0.83-0.99]). In animal models, the lovastatin group had significantly reduced infarct volume, decreased immobility time and latency to eat, a reduced number of Fluoro- Jade-positive cells and levels of glial fibrillary acidic protein and tumor necrosis factor-alpha, and increased adenosine monophosphate -activated protein kinase (AMPK) and its upstream kinase liver kinase B1 in the hippocampal dentate gyrus. These effects were blocked in AMPK inhibitor-pretreated TBI rats. CONCLUSIONS: Our epidemiological data showed that a decreased risk of depression was associated with statin pretreatment, which was supported by an animal study. The underlying mechanism for this appears to involve AMPK activation in the statin pretreatment-induced alleviation of TBI.


Subject(s)
Brain Injuries, Traumatic , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Humans , Rats , Animals , Lovastatin/pharmacology , Lovastatin/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Depression/drug therapy , Depression/etiology , AMP-Activated Protein Kinases/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism
20.
Ultrasonics ; 131: 106949, 2023 May.
Article in English | MEDLINE | ID: mdl-36773481

ABSTRACT

The meningeal lymphatic system drains the cerebrospinal fluid from the subarachnoid space to the cervical lymphatic system, primarily to the deep cervical lymph nodes. Perturbations of the meningeal lymphatic system have been linked to various neurologic disorders. A method to specifically monitor the flow of meningeal lymphatic system in real time is unavailable. In the present study, we adopted the high-frequency ultrasound (HFUS) with 1,1'diocatadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI)-loaded microbubble and FePt@PLGA nanoparticle contrast agents to evaluate the flow of the meningeal lymphatic system in 2-month-old mice. Statistical analysis was performed to identify changes of HFUS signals among the microbubbles, FePt@PLGA nanoparticles, and saline control groups. Approximately 15 min from the start of intracerebroventricular injection of contrast agents, their signals were evident at the deep cervical lymph nodes and lasted for at least 60 min. These signals were validated on the basis of the presence of DiI and Fe signals in the deep cervical lymph nodes. Ligation of afferent lymphatic vessels to the deep cervical lymph nodes eliminated the HFUS signals. Moreover, ablation of lymphatic vessels near the confluence of sinuses decreased the HFUS signals in the deep cervical lymph nodes. Glioma-bearing mice that exhibited reduced lymphatic vessel immunostaining signals near the confluence of sinuses had lowered HFUS signals in the deep cervical lymph nodes within 60 min. The proposed method provides a minimally invasive approach to monitor the qualities of the meningeal lymphatic system in real time as well as the progression of the meningeal lymphatic system in various brain disease animal models.


Subject(s)
Lymph Nodes , Lymphatic Vessels , Mice , Animals , Lymph Nodes/pathology , Contrast Media , Lymphatic System/diagnostic imaging , Lymphatic Vessels/diagnostic imaging , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL