Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Brain Res Bull ; 185: 64-73, 2022 07.
Article in English | MEDLINE | ID: mdl-35489671

ABSTRACT

Reconsolidation of drug memories is the process of restoring unstable memories after unconditioned (UCS; e.g., drugs) or conditioned stimulus (CS; e.g., drug-paired contexts), and provides promise for prevention of drug relapse. Circular RNAs (circRNAs) have important effects on the transcription and post-transcriptional regulation of gene expression. However, the role of circRNAs in the reconsolidation of drug memories is unclear. Here, we observed that cocaine-induced memory retrieval significantly increased circTmeff-1 level in the nucleus accumbens (NAc) core but not shell. Importantly, the disrupted expression of circTmeff-1 using virus in the NAc core damaged the reconsolidation of cocaine-associated memories. The knockdown of circTmeff-1 in the NAc shell or without UCS retrieval or 9 h after UCS retrieval had no such effects. Mechanistically, using bioinformatic analysis and loss- or gain- of function assays, we revealed that antagomiR-206 reversed the inhibitory effect of circTmeff-1 knockdown on the expression of brain-derived neurotrophic factor (BDNF) during the reconsolidation of cocaine-associated memories. Taken together, these results demonstrate the role of circTmeff-1 in the reconsolidation of cocaine-associated memory and that circTmeff-1 may function as a decoy for miR-206 to regulate the expression of BDNF.


Subject(s)
Cocaine , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cocaine/pharmacology , Nucleus Accumbens , RNA, Circular , Rats , Rats, Sprague-Dawley
2.
Pharmacol Res ; 170: 105722, 2021 08.
Article in English | MEDLINE | ID: mdl-34116208

ABSTRACT

A progressive increase in drug craving following drug exposure is an important trigger of relapse. CircularRNAs (CircRNAs), key regulators of gene expression, play an important role in neurological diseases. However, the role of circRNAs in drug craving is unclear. In the present study, we trained mice to morphine conditioned place preference (CPP) and collected the nucleus accumbens (NAc) sections on abstinence day 1 (AD1) and day 14 (AD14) for RNA-sequencing. CircTmeff-1, which was highly expressed in the NAc core, was associated with incubation of context-induced morphine craving. The gain- and loss- of function showed that circTmeff-1 was a positive regulator of incubation. Simultaneously, the expression of miR-541-5p and miR-6934-3p were down-regulated in the NAc core during the incubation period. The dual luciferase reporter, RNA pulldown, and fluorescence insitu hybridization assays confirmed that miR-541-5p and miR-6934-3p bind to circTmeff-1 selectively. Furthermore, bioinformatics and western blot analysis suggested that vesicle-associated membrane protein 1 (VAMP1) and neurofascin (NFASC), both overlapping targets of miR-541-5p and miR-6934-3p, were highly expressed during incubation. Lastly, AAV-induced down-regulation of circTmeff-1 decreased VAMP1 and NFASC expression and incubation of morphine craving. These findings suggested that circTmeff-1, a novel circRNA, promotes incubation of context-induced morphine craving by sponging miR-541/miR-6934 in the NAc core. Thus, circTmeff-1 represents a potential therapeutic target for context-induced opioid craving, following prolonged abstinence.


Subject(s)
Behavior, Animal , Craving , Drug-Seeking Behavior , Morphine Dependence/metabolism , Nucleus Accumbens/metabolism , RNA, Circular/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cues , Disease Models, Animal , Gene Expression Regulation , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Morphine Dependence/genetics , Morphine Dependence/physiopathology , Morphine Dependence/psychology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nucleus Accumbens/physiopathology , RNA, Circular/genetics , Vesicle-Associated Membrane Protein 1/genetics , Vesicle-Associated Membrane Protein 1/metabolism
3.
Brain Res Bull ; 162: 73-83, 2020 09.
Article in English | MEDLINE | ID: mdl-32544512

ABSTRACT

Methamphetamine (METH) is an illegal amphetamine-typed psychostimulant that is abused worldwide and causes serious public health problems. METH exposure induces apoptosis and autophagy in neuronal cells. However, the role of pyroptosis in METH-induced neurotoxicity is still unclear. Here, we investigate whether pyroptosis is involved in METH-induced hippocampal neurotoxicity and the potential mechanisms of Endoplasmic reticulum (ER) stress in hippocampal neuronal cells. For this purpose, the expression levels of pyroptosis-related proteins, GSDMD and GSDME, were analyzed by immunoblotting and immunohistochemistry in the hippocampal neuron cell line HT-22. Next, we explored METH-induced pyroptosis in HT-22 using immunoblotting, LDH assays and SYTOX green acid staining. Further, the relationship between pyroptosis and ER stress in METH-induced hippocampal neuron damage was studied in HT-22 cells using inhibitors including TUDCA, a specific inhibitor of ER stress, GSK-2656157, a PERK pathway inhibitor and STF-0803010, an inhibitor of IRE1α endoribonuclease activity. This relationship was also studied using siRNAs, including siTRAF2, an siRNA against IRE1α kinase activity and siATF6 against the ATF6 pathway, which were analyzed by immunoblotting, LDH assays and SYTOX green acid staining. GSDME but not GSDMD was found to be expressed in HT-22 cells. METH treatment induced the upregulation of cleaved GSDME-NT and LDH release, as well as the increase of SYTOX green positive cells in HT-22 cells, which was partly reversed by inhibitors and siRNAs, indicating that the ER stress signaling pathway was involved in GSDME-dependent cell death induced by METH. In summary, these results revealed that METH induced ER stress that mediated GSDME-dependent cell death in hippocampal neuronal cells. These findings provide novel insight into the mechanisms of METH-induced neurotoxicity.


Subject(s)
Central Nervous System Stimulants/pharmacology , Endoplasmic Reticulum Stress/physiology , Hippocampus/metabolism , Methamphetamine/pharmacology , Neurons/metabolism , Receptors, Estrogen/biosynthesis , Animals , Cell Death/drug effects , Cell Death/physiology , Cell Line , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Hippocampus/drug effects , Mice , Neurons/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
4.
Front Pharmacol ; 8: 639, 2017.
Article in English | MEDLINE | ID: mdl-28959203

ABSTRACT

Methamphetamine (METH) abuse causes serious health problems worldwide, and long-term use of METH disrupts the blood-brain barrier (BBB). Herein, we explored the potential mechanism of endoplasmic reticulum (ER) stress in METH-induced BBB endothelial cell damage in vitro and the therapeutic potential of endoplasmic reticulum stress inhibitors for METH-induced BBB disruption in C57BL/6J mice. Exposure of immortalized BMVEC (bEnd.3) cells to METH significantly decreased cell viability, induced apoptosis, and diminished the tightness of cell monolayers. METH activated ER stress sensor proteins, including PERK, ATF6, and IRE1, and upregulated the pro-apoptotic protein CHOP. The ER stress inhibitors significantly blocked the upregulation of CHOP. Knockdown of CHOP protected bEnd.3 cells from METH-induced cytotoxicity. Furthermore, METH elevated the production of reactive oxygen species (ROS) and induced the dysfunction of mitochondrial characterized by a Bcl2/Bax ratio decrease, mitochondrial membrane potential collapse, and cytochrome c. ER stress release was partially reversed by ROS inhibition, and cytochrome c release was partially blocked by knockdown of CHOP. Finally, PBA significantly attenuated METH-induced sodium fluorescein (NaFluo) and Evans Blue leakage, as well as tight junction protein loss, in C57BL/6J mice. These data suggest that BBB endothelial cell damage was caused by METH-induced endoplasmic reticulum stress, which further induced mitochondrial dysfunction, and that PBA was an effective treatment for METH-induced BBB disruption.

5.
Neuropharmacology ; 118: 199-208, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28359771

ABSTRACT

Hydrogen therapy is a new medical approach for a wide range of diseases. The effects of hydrogen on central nervous system-related diseases have recently become increasingly appreciated, but little is known about whether hydrogen affects the morphine withdrawal process. This study aims to investigate the potential effects of hydrogen-rich saline (HRS) administration on naloxone-precipitated withdrawal symptoms and morphine withdrawal-induced anxiety-like behaviors. Mice received gradually increasing doses (25-100 mg/kg, i.p.) of morphine over 3 days. In the naloxone-precipitated withdrawal procedure, the mice were treated with three HRS (20 µg/kg, i.p.) injections, and naloxone (1 mg/kg, i.p.) was given 30 min after HRS administration. Body weight, jumping behavior and wet-dog shakes were immediately assessed. In the spontaneous withdrawal procedure, the mice were treated with HRS (20 µg/kg, i.p.) every 8-h. Mice underwent naloxone-precipitated or spontaneous withdrawal were tested for anxiety-like behaviors in the elevated plus-maze (EPM) and light/dark box (L/D box) paradigm, respectively. In addition, the levels of plasma corticosterone were measured. We found that HRS administration significantly reduced body weight loss, jumping behavior and wet-dog shakes in mice underwent naloxone-precipitated withdrawal, and attenuated anxiety-like behaviors in the EPM and L/D box tests after naloxone-precipitated withdrawal or a 2-day spontaneous withdrawal period. Hypo-activity or motor impairment after HRS administration was not observed in the locomotion tests. Furthermore, HRS administration significantly decreased the levels of corticosterone in morphine-withdrawn mice. These are the first findings to indicate that hydrogen might ameliorate withdrawal symptoms and exert an anxiolytic-like effect in morphine-withdrawal mice.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Anxiety/etiology , Hydrogen/therapeutic use , Morphine/adverse effects , Substance Withdrawal Syndrome/complications , Adaptation, Ocular/drug effects , Animals , Corticosterone/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Head Movements/drug effects , Hydrocortisone/blood , Locomotion/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Naloxone/therapeutic use , Sodium Chloride/therapeutic use , Statistics, Nonparametric , Substance Withdrawal Syndrome/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...