Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Addict Biol ; 25(5): e12808, 2020 09.
Article in English | MEDLINE | ID: mdl-31364211

ABSTRACT

Cocaine addiction is a chronic relapsing brain disorder characterized by compulsive drug seeking. Preliminary study suggested that bromodomain-containing protein 4 (BRD4), an epigenetic reader protein, participates in cocaine-induced reward and neuroplasticity. However, the exact role of BRD4 in cocaine addiction, particularly cocaine relapse, remains elusive. In this study, we found that BRD4 phosphorylation in the nucleus accumbens (NAc) was closely related to the maintenance of cocaine reinforcement and relapse in different cocaine exposure paradigms. Cocaine significantly increased the binding of phosphorylated BRD4 (pBRD4) at the promoter of Gria2 and Bdnf genes in the NAc. (+)JQ1, a selective BRD4 inhibitor, markedly reduced the reinforcement and reinstatement of cocaine-seeking behaviors, which was accompanied by the decreased expressions of GRIA2 and BDNF. Furthermore, chromatin immunoprecipitation assay showed that (+)JQ1 clearly attenuated cocaine-enhanced binding of pBRD4 at the promotor of Gria2 and Bdnf genes. Blockade of casein kinase II significantly attenuated BRD4 phosphorylation and cocaine relapse-like behaviors, suggesting the important role of pBRD4 in modulating cocaine effect. Together, our findings suggest that BRD4 phosphorylation in the NAc modulates multiple addiction-related behaviors of cocaine and particularly relapse to cocaine-seeking behaviors. Inhibition of BRD4 activity may be a novel target against cocaine addiction and relapse.


Subject(s)
Cocaine-Related Disorders/genetics , Cocaine/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleus Accumbens/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Conditioning, Operant , Disease Models, Animal , Dopamine Uptake Inhibitors/pharmacology , Drug-Seeking Behavior/drug effects , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Recurrence , Self Administration
2.
Transl Psychiatry ; 9(1): 199, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434869

ABSTRACT

Circular RNA (circRNA), a novel type of endogenous non-coding RNA, plays natural miRNA sponge effect that represses the activities of corresponding miRNAs through binding with them, thus modulating transcriptional expression of genes. Recent studies indicate that circRNAs are significantly enriched in the brain and some of them are derived from synaptic protein-coding genes. In addition, miRNAs are involved in synaptic plasticity, memory formation, and cocaine addiction. However, the role of circRNAs in cocaine reward is unclear. This study aimed to investigate the expression profile of striatal circRNAs in the mice after cocaine self-administration. By using circRNA microarray analysis, we observed that 90 striatal circRNAs were differentially expressed in cocaine self-administering mice, of which 18 circRNAs were up-regulated and 72 down-regulated. Six circRNAs were selected randomly for validation by using quantitative reverse transcription-PCR, and their expression levels showed consistency with microarray analysis. We backward predicted the circRNAs and their binding sites of miRNAs associated with neuroplasticity. In functional validation test, mmu_circRNA_002381 may modulate the transcription of certain genes associated with neuroplasticity, such as limk1 and bdnf. Taken together, circRNAs may participate in cocaine behavioral effect via interacting with miRNAs. Our findings reveal a potential role of circRNAs in cocaine effect.


Subject(s)
Cocaine/pharmacology , Corpus Striatum/drug effects , Dopamine Uptake Inhibitors/pharmacology , Gene Expression Regulation/drug effects , RNA, Circular/metabolism , Animals , Corpus Striatum/metabolism , Down-Regulation/drug effects , Male , Mice , RNA, Circular/genetics , Self Administration , Up-Regulation/drug effects
3.
Neuropharmacology ; 148: 377-393, 2019 04.
Article in English | MEDLINE | ID: mdl-28987936

ABSTRACT

The brain is a high energy-consuming organ that typically utilizes glucose as the main energy source for cerebral activity. When glucose becomes scarce under conditions of stress, ketone bodies, such as ß-hydroxybutyrate, acetoacetate and acetone, become extremely important. Alterations in brain energy metabolism have been observed in psychostimulant abusers; however, the mode of brain metabolic programming in cocaine dependence remains largely unknown. Here, we profiled the metabolites and metabolic enzymes from brain nucleus accumbens (NAc) of mice exposed to cocaine. We found that cocaine modified energy metabolism and markedly activated ketogenesis pathway in the NAc. The expression of HMG-CoA synthase 2 (HMGCS2), a critical rate-limiting ketogenesis enzyme, was markedly up-regulated. After switching metabolic pathways from ketogenesis to glycolysis through activation of glucokinase, cocaine-evoked metabolic reprogramming regained homeostasis, and the cocaine effect was attenuated. Importantly, both the pharmacological and genetic inhibition of HMGCS2 significantly suppressed cocaine-induced ketogenesis and behavior. In conclusion, cocaine induces a remarkable energy reprogramming in the NAc, which is characterized by HMGCS2-driven ketogenesis. Such effect may facilitate adaptations to cocaine-induced energy stress in the brain. Our findings establish an important link between drug-induced energy reprogramming and cocaine effect, and may have implication in the treatment of cocaine addiction.


Subject(s)
Cocaine/pharmacology , Energy Metabolism/drug effects , Hydroxymethylglutaryl-CoA Synthase/biosynthesis , Ketone Bodies/metabolism , Animals , Homeostasis , Hydroxymethylglutaryl-CoA Synthase/antagonists & inhibitors , Male , Mice , Nucleus Accumbens/metabolism , Up-Regulation/drug effects
4.
Toxicol Lett ; 294: 166-176, 2018 Sep 15.
Article in English | MEDLINE | ID: mdl-29758358

ABSTRACT

Alcohol abuse and its related diseases are the major risk factors for human health. Although the mechanism of alcohol-related disorders has been widely investigated, serum metabolites associated with long-term alcohol intake have not been well explored. In this study, we aimed to investigate the profiles of serum metabolites and lipid species of rats chronically exposed to alcohol, which may be involved in the pathogenesis of alcohol-associated disease. An 1H NMR-based metabolomics and Q-TOF/MS-based lipidomics approach were applied to investigate the profile of serum metabolites and lipid species of rats administrated daily with alcohol (12% vol/vol, 10 ml/kg per day, i.g.) for one year continuously. The rats administered with sterile water (10 ml/kg per day, i.g.) were used as control. We found that alcohol affected mostly the lipid species rather than small molecule metabolites in the serum of both female and male rats. Among the modified lipids, glycerophospholipid, sphingolipid and glycerolipids metabolism pathways were profoundly altered. The prominent changes in lipid profiles included diacylglycerol (DG), lysophosphatidylcholine (LysoPC), phosphatidic acid (PA), phosphatidylcholine (PC), phosphatidylethanolamine (PE) and triacylglycerol (TG). Moreover, fatty-acyl profile of lipids and total degree of unsaturation of fatty acid were also significantly altered by alcohol. The modified lipidomic profile may help to understand the pathogenesis of alcohol-associated diseases and also be of value for clinical evaluation of alcohol abuse, alcohol-associated disease diagnosis.


Subject(s)
Alcoholism/physiopathology , Disease Models, Animal , Dyslipidemias/etiology , Lipids/blood , Alcoholism/blood , Animals , Cholesterol/blood , Cholesterol/chemistry , Discriminant Analysis , Fatty Acids/analysis , Fatty Acids/blood , Fatty Acids/chemistry , Female , Glycerides/blood , Glycerides/chemistry , Glycerophospholipids/blood , Glycerophospholipids/chemistry , Least-Squares Analysis , Lipids/chemistry , Magnetic Resonance Spectroscopy , Male , Metabolomics/methods , Molecular Structure , Random Allocation , Rats, Wistar , Reproducibility of Results , Sex Characteristics , Spectrometry, Mass, Electrospray Ionization , Sphingolipids/blood , Sphingolipids/chemistry
5.
Exp Neurol ; 307: 52-61, 2018 09.
Article in English | MEDLINE | ID: mdl-29753648

ABSTRACT

Nicotinamide phosphoribosyltransferase (NAMPT), a rate-limiting enzyme in nicotinamide adenine dinucleotide (NAD) biosynthesis in mammals, converts nicotinamide into nicotinamide mononucleotide (NMN). NMN is subsequently converted to NAD, a component that is critical for cell energy metabolism and survival. Sirtuin 1 (SIRT1), an NAD-dependent histone deacetylase, plays an important role in mediating memory and synaptic plasticity. Here, we found that NAMPT was significantly upregulated in the ventral tegmental area (VTA) of cocaine-conditioned mice. Intraperitoneal or intra-VTA injection of FK866, a specific inhibitor of NAMPT, significantly attenuated cocaine reward. However, such effects were clearly repressed by intra-VTA expression of NAMPT or supplementation with NMN. Using 1H-nuclear magnetic resonance metabolomic analysis, we found that the content of NAD and NMN were increased in the VTA of cocaine-conditioned mice; moreover, the expression of SIRT1 was also upregulated. Interestingly, the inhibitory effect of FK866 on cocaine reward was significantly weakened in Sirt1 midbrain conditional knockout mice. Our results suggest that NAMPT-mediated NAD biosynthesis may modify cocaine behavioral effects through SIRT1. Moreover, our findings reveal that the interplay between NAD biosynthesis and SIRT1 regulation may comprise a novel regulatory pathway that responds to chronic cocaine stimuli.


Subject(s)
Cocaine/pharmacology , Cytokines/biosynthesis , Nicotinamide Phosphoribosyltransferase/biosynthesis , Reward , Sirtuin 1/biosynthesis , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dopamine Uptake Inhibitors/pharmacology , Locomotion/drug effects , Locomotion/physiology , Magnetic Resonance Spectroscopy/methods , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
6.
Neurotherapeutics ; 15(2): 500-510, 2018 04.
Article in English | MEDLINE | ID: mdl-29464572

ABSTRACT

Methamphetamine (METH) abuse has become a major public health concern worldwide without approved pharmacotherapies. The brain renin-angiotensin system (RAS) is involved in the regulation of neuronal function as well as neurological disorders. Angiotensin II (Ang II), which interacts with Ang II type 1 receptor (AT1-R) in the brain, plays an important role as a neuromodulator in dopaminergic transmission. However, the role of brain RAS in METH-induced behavior is largely unknown. Here, we revealed that repeated METH administration significantly upregulated the expression of AT1-R in the striatum of mice, but downregulated dopamine D3 receptor (D3R) expression. A specific AT1-R blocker telmisartan, which can penetrate the brain-blood barrier (BBB), or genetic deletion of AT1-R was sufficient to attenuate METH-triggered hyperlocomotion in mice. However, intraperitoneal injection of AT1-R blocker losartan, which cannot penetrate BBB, failed to attenuate METH-induced behavior. Moreover, intra-striatum re-expression of AT1 with lentiviral virus expressing AT1 reversed the weakened locomotor activity of AT1-/- mice treated with METH. Losartan alleviated METH-induced cytotoxicity in SH-SY5Y cells in vitro, which was accompanied by upregulated expressions of D3R and dopamine transporter. In addition, intraperitoneal injection of perindopril, which is a specific ACE inhibitor and can penetrate BBB, significantly attenuated METH-induced hyperlocomotor activity. Collectively, our results show that blockade of brain RAS attenuates METH-induced hyperlocomotion and neurotoxicity possibly through modulation of D3R expression. Our findings reveal a novel role of Ang II-AT1-R in METH-induced hyperlocomotion.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Corpus Striatum/physiopathology , Hyperkinesis/physiopathology , Methamphetamine/administration & dosage , Methamphetamine/toxicity , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/drug effects , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Animals , Cell Line, Tumor , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Humans , Hyperkinesis/chemically induced , Losartan/administration & dosage , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor, Angiotensin, Type 1/genetics , Receptors, Dopamine D3/metabolism , Telmisartan/administration & dosage , Up-Regulation
7.
Toxicol Lett ; 279: 67-76, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28689763

ABSTRACT

Lipids are predominant components of the brain and key regulators for neural structure and function. The effect of methamphetamine (METH) on behavior, cognition as well as memory has been intensively investigated; however, the impact of METH on brain lipid profiles is largely unknown. Here, we used a global lipidomic approach to investigate brain lipidome of METH-sensitized mice. We found that repeated METH significantly modified the lipidome in the hippocampus, prefrontal cortex (PFC) and striatum. Interestingly, nucleus accumbens showed no obvious alteration in lipidomic profiling. Phospholipid and sphingolipid metabolisms were profoundly modified in the hippocampus of METH-sensitized mice, exhibiting increased phosphatidic acid and ether phosphatidylcholine but decreased lysophosphatidylethanolamine, lactosylceramide and triglycerides. The fatty acyl length of phospholipids and diacylglycerol longer than 40 carbon were clearly decreased in the hippocampus, and that 36 carbon was decreased in the PFC. These results indicate METH can profoundly affect the metabolism of phospholipids, sphingolipids and glycerolipids in the brain. Our findings reveal a link between remodeled brain lipidome and neurobehavior induced by METH.


Subject(s)
Basal Ganglia/drug effects , Central Nervous System Stimulants/toxicity , Hippocampus/drug effects , Lipid Metabolism/drug effects , Methamphetamine/toxicity , Prefrontal Cortex/drug effects , Animals , Basal Ganglia/metabolism , Behavior, Animal/drug effects , Central Nervous System Stimulants/administration & dosage , Fatty Acids/metabolism , Glycerides/metabolism , Hippocampus/metabolism , Male , Methamphetamine/administration & dosage , Mice, Inbred C57BL , Motor Activity/drug effects , Phospholipids/metabolism , Prefrontal Cortex/metabolism , Sphingolipids/metabolism , Time Factors
8.
Neuropsychopharmacology ; 42(12): 2333-2343, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28726800

ABSTRACT

Nicotinamide N-methyltransferase (NNMT) transfers the methyl from S-adenosyl-L-methionine (SAM) to nicotinamide (NA) to produce S-adenosyl-L-homocysteine (SAH) and 1-methylnicotinamide (MeN). NNMT has been implicated in a variety of diseases; however, the role of NNMT in drug addiction is largely unknown. Here, we found that the expression of Nnmt was significantly upregulated in the dorsal striatum (DS) of cocaine-conditioned mice. Cocaine significantly decreased SAM/SAH ratio levels in the DS, which was accompanied with the decreased activities of Rac1 and RhoA. Lentivirus-mediated knockdown of Nnmt in the dorsomedial striatum (DMS) attenuated cocaine conditioned place preference (CPP) reward, but increased striatal SAM/SAH ratio levels as well as Rac1 and RhoA activities. In addition, pharmacological inhibition of NNMT through intra-DMS infusion of MeN attenuated cocaine CPP and the activities of Rac1 and RhoA, but increased SAM/SAH ratio. These results suggest that NNMT-dependent transmethylation is involved in the activation of Rac1 and RhoA, which utilize SAM as a methyl donor cofactor. Co-immunoprecipitation assay using a RhoGDIα antibody indirectly captured Rac1 or RhoA that were bound to RhoGDIα. The results showed that cocaine increased the association of RhoGDIα with Rac1 or RhoA, whereas such effect was inhibited by Nnmt knockdown. Collectively, our findings show that NNMT regulates cocaine CPP through SAM-mediated modification of Rac1 and RhoA.


Subject(s)
Cocaine/administration & dosage , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Nicotinamide N-Methyltransferase/biosynthesis , Animals , Drug Administration Schedule , Male , Mice , Mice, Inbred C57BL
9.
Neurotox Res ; 28(4): 290-301, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26233726

ABSTRACT

Sorafenib, an active multi-kinase inhibitor, has been widely used as a chemotherapy drug to treat advanced clear-cell renal cell carcinoma patients. In spite of the relative safety, sorafenib has been shown to exert a negative impact on cognitive functioning in cancer patients, specifically on learning and memory; however, the underlying mechanism remains unclear. In this study, an NMR-based metabolomics approach was applied to investigate the neurochemical effects of sorafenib in rats. Male rats were once daily administrated with 120 mg/kg sorafenib by gavage for 3, 7, and 28 days, respectively. NMR-based metabolomics coupled with histopathology examinations for hippocampus, prefrontal cortex (PFC), and striatum were performed. The (1)H NMR spectra data were analyzed by using multivariate pattern recognition techniques to show the time-dependent biochemical variations induced by sorafenib. Excellent separation was obtained and distinguishing metabolites were observed between sorafenib-treated and control rats. A total of 36 differential metabolites in hippocampus of rats treated with sorafenib were identified, some of which were significantly changed. Furthermore, these modified metabolites mainly reflected the disturbances in neurotransmitters, energy metabolism, membrane, and amino acids. However, only a few metabolites in PFC and striatum were altered by sorafenib. Additionally, no apparent histological changes in these three brain regions were observed in sorafenib-treated rats. Together, our findings demonstrate the disturbed metabonomics pathways, especially, in hippocampus, which may underlie the sorafenib-induced cognitive deficits in patients. This work also shows the advantage of NMR-based metabolomics over traditional approach on the study of biochemical effects of drugs.


Subject(s)
Brain/metabolism , Metabolomics , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Amino Acids/metabolism , Animals , Antioxidants/metabolism , Cell Membrane/metabolism , Energy Metabolism , Magnetic Resonance Imaging , Male , Niacinamide/administration & dosage , Rats , Rats, Sprague-Dawley , Sorafenib
10.
Neurobiol Dis ; 82: 54-65, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26054440

ABSTRACT

Studies have showed that prenatal cocaine exposure (PCOC) can impair cognitive function and social behavior of the offspring; however, the mechanism underlying such effect is poorly understood. Insulin-like growth factor II (Igf-II), an imprinted gene, has a critical role in memory consolidation and enhancement. We hypothesized that epigenetic regulation of hippocampal Igf-II may attribute to the cognitive deficits of PCOC offspring. We used Morris water maze and open-field task to test the cognitive function in PCOC offspring. The epigenetic alteration involved in hippocampal Igf-II expression deficit in PCOC offspring was studied by determining Igf-II methylation status, DNA methyltransferases (DNMT) expressions and L-methionine level. Moreover, IGF-II rescue experiments were performed and the downstream signalings were investigated in PCOC offspring. In behavioral tests, we observed impaired spatial learning and memory and increased anxiety in PCOC offspring; moreover, hippocampal IGF-II mRNA and protein expressions were significantly decreased. Hippocampal methylation of cytosine-phospho-guanine (CpG) dinucleotides in differentially methylated region (DMR) 2 of Igf-II was elevated in PCOC offspring, which may be driven by the upregulation of L-methionine and DNA methyltransferase (DNMT) 1. Importantly, intra-hippocampal injection of recombinant IGF-II reactivated the repressed calcium calmodulin kinase II α (CaMKIIα) and reversed cognitive deficits in PCOC offspring. Collectively, our findings suggest that cocaine exposure during pregnancy impairs cognitive function of offspring through epigenetic modification of Igf-II gene. Enhancing IGF-II signaling may represent a novel therapeutical strategy for cocaine-induced cognitive impairment.


Subject(s)
Cocaine/pharmacology , Cognition/drug effects , Epigenesis, Genetic , Insulin-Like Growth Factor II/genetics , Prenatal Exposure Delayed Effects/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cognition/physiology , DNA Methylation/drug effects , Female , Hippocampus/drug effects , Hippocampus/metabolism , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor II/pharmacology , Maze Learning/drug effects , Maze Learning/physiology , Mice , Phosphorylation , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...