Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Behav Pharmacol ; 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38847447

ABSTRACT

Excessive prescribing and misuse of prescription opioids, such as oxycodone, significantly contributed to the current opioid crisis. Although oxycodone is typically consumed orally by humans, parenteral routes of administration have primarily been used in preclinical models of oxycodone dependence. To address this issue, more recent studies have used oral self-administration procedures to study oxycodone seeking and withdrawal in rodents. Behavioral differences, however, following oral oxycodone intake versus parenteral oxycodone administration remain unclear. Thus, the goal of the current studies was to compare anxiety- and withdrawal-like behaviors using established opioid dependence models of either home cage oral intake of oxycodone (0.5 mg/ml) or repeated subcutaneous (s.c.) injections of oxycodone (10 mg/kg) in male and female mice. Here, mice received 10 days of oral or s.c. oxycodone administration, and following 72 h of forced abstinence, anxiety- and withdrawal-like behaviors were measured using elevated zero maze, open field, and naloxone-induced precipitated withdrawal procedures. Global withdrawal scores were increased to a similar degree following oral and s.c. oxycodone use, while both routes of oxycodone administration had minimal effects on anxiety-like behaviors. When examining individual withdrawal-like behaviors, mice receiving s.c. oxycodone exhibited more paw tremors and jumps during naloxone-induced precipitated withdrawal compared with oral oxycodone mice. These results indicate that both models of oxycodone administration are sufficient to elevate global withdrawal scores, but, when compared with oral consumption, s.c. oxycodone injections yielded more pronounced effects on some withdrawal-like behaviors.

2.
Drug Alcohol Depend ; 253: 110987, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37864957

ABSTRACT

Despite the thousands of lives lost during the ongoing opioid crisis, a scarcity of new and effective clinical treatments for opioid use disorder (OUD) remains. To address this unmet need, some researchers have turned to dissociative and psychedelic drugs to treat multiple psychiatric conditions. In particular, low doses of ketamine have been shown to attenuate opioid withdrawal and drug use in clinical and preclinical studies. However, ketamine has misuse liability and dissociative side effects that may limit its widespread application as a treatment for OUD. More recently, (2R,6R)-hydroxynorketamine (HNK), a ketamine metabolite that lacks misuse potential, has gained attention for its effectiveness in depression and stress models. To uncover its role in OUD, we tested the time-dependent effects of (2R,6R)-HNK on oxycodone withdrawal and reinstatement of oxycodone conditioned place preference (CPP). In male and female oxycodone-dependent mice, we found that 24h pretreatment with (2R,6R)-HNK (10 or 30mg/kg, s.c.) reduced the frequency of withdrawal-like behaviors and global withdrawal scores during naloxone-precipitated withdrawal, whereas 1h pretreatment with (2R,6R)-HNK only reduced paw tremors and the sum of global withdrawal scores but not GWS Z-scores. In other experiments, both 1h and 24h pretreatment with (2R,6R)-HNK (30mg/kg, s.c.) blocked drug-induced reinstatement of oxycodone CPP. Finally, we found (2R,6R)-HNK (30mg/kg, sc) had no effect on locomotor activity and thigmotaxis. Together, these results indicate that acute (2R,6R)-HNK has efficacy in some preclinical models of OUD without producing locomotor or anxiety-like side effects.


Subject(s)
Hallucinogens , Ketamine , Humans , Mice , Male , Female , Animals , Ketamine/pharmacology , Antidepressive Agents , Oxycodone/pharmacology , Oxycodone/therapeutic use
3.
Neuropsychopharmacology ; 47(8): 1431-1439, 2022 07.
Article in English | MEDLINE | ID: mdl-35351961

ABSTRACT

Innovative breakthroughs in nanotechnology are having a substantial impact in healthcare, especially for brain diseases where effective therapeutic delivery systems are desperately needed. Nanoparticle delivery systems offer an unmatched ability of not only conveying a diverse array of diagnostic and therapeutic agents across complex biological barriers, but also possess the ability to transport payloads to targeted cell types over a sustained period. In substance use disorder (SUD), many therapeutic targets have been identified in preclinical studies, yet few of these findings have been translated to effective clinical treatments. The lack of success is, in part, due to the significant challenge of delivering novel therapies to the brain and specific brain cells. In this review, we evaluate the potential approaches and limitations of nanotherapeutic brain delivery systems. We also highlight the examples of promising strategies and future directions of nanocarrier-based treatments for SUD.


Subject(s)
Nanoparticles , Substance-Related Disorders , Brain/metabolism , Drug Delivery Systems , Humans , Nanoparticles/therapeutic use , Nanotechnology , Substance-Related Disorders/drug therapy , Substance-Related Disorders/metabolism
4.
Neuropharmacology ; 210: 109040, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35314160

ABSTRACT

Epigenetic pharmacotherapies have emerged as a promising treatment option for substance use disorder (SUD) due to their ability to reverse maladaptive transcriptional and behavioral responses to drugs of abuse. In particular, inhibitors of bromodomain and extra terminal domain (BET) reader proteins have been shown to reduce cocaine- and opioid-seeking behaviors in rodents. However, only pan-BET inhibitors, small molecules that bind to both bromodomains (BD1 and BD2) with all BET proteins, have been investigated in animal models of SUD. Given the potential side effects associated with pan-BET inhibitors, safer and more selective strategies are needed to advance BET therapeutics as a potential treatment for SUD. Here, we show that RVX-208, a clinically tested, BD2-selective BET inhibitor, dose-dependently reduced cocaine conditioned place preference in male and female mice, similar to the pan-BET inhibitor JQ1. In other behavioral experiments, RVX-208 treatment did not alter distance traveled, anxiety-like behavior, or novel object recognition memory. At the transcriptional level, RVX-208 attenuated the expression of multiple cocaine-induced genes in the nucleus accumbens in a sex-dependent manner. RVX-208 produced a distinct transcriptional response in stimulated primary neurons compared to JQ1 but had little effect on gene expression in non-stimulated neurons. Together, these data indicate that targeting domain-specific BET mechanisms may be an effective and safer strategy to reduce cocaine-induced neurobehavioral adaptations.


Subject(s)
Cocaine , Animals , Cocaine/pharmacology , Epigenomics , Female , Male , Mice , Protein Domains
5.
Article in English | MEDLINE | ID: mdl-36601439

ABSTRACT

Although noncoding RNAs (ncRNAs) have been shown to regulate maladaptive neuroadaptations that drive compulsive drug use, ncRNA-targeting therapeutics for substance use disorder (SUD) have yet to be clinically tested. Recent advances in RNA-based drugs have improved many therapeutic issues related to immune response, specificity, and delivery, leading to multiple successful clinical trials for other diseases. As the need for safe and effective treatments for SUD continues to grow, novel nucleic acid-based therapeutics represent an appealing approach to target ncRNA mechanisms in SUD. Here, we review ncRNA processes implicated in SUD, discuss recent therapeutic approaches for targeting ncRNAs, and highlight potential opportunities and challenges of ncRNA-targeting therapeutics for SUD.

6.
Neuropharmacology ; 181: 108306, 2020 12 15.
Article in English | MEDLINE | ID: mdl-32946883

ABSTRACT

Epigenetic pharmacotherapy for CNS-related diseases is a burgeoning area of research. In particular, members of the bromodomain and extra-terminal domain (BET) family of proteins have emerged as intriguing therapeutic targets due to their putative involvement in an array of brain diseases. With their ability to bind to acetylated histones and act as a scaffold for chromatin modifying complexes, BET proteins were originally thought of as passive epigenetic 'reader' proteins. However, new research depicts a more complex reality where BET proteins act as key nodes in lineage-specific and signal-dependent transcriptional mechanisms to influence disease-relevant functions. Amid a recent wave of drug development efforts from basic scientists and pharmaceutical companies, BET inhibitors are currently being studied in several CNS-related disease models, but safety and tolerability remain a concern. Here we review the progress in understanding the neurobiological mechanisms of BET proteins and the therapeutic potential of targeting BET proteins for brain health and disease.


Subject(s)
Brain Diseases/genetics , Brain/physiology , Epigenomics , Protein Domains/genetics , Animals , Brain Diseases/therapy , Genetic Therapy , Humans
7.
Biochem Pharmacol ; 168: 269-274, 2019 10.
Article in English | MEDLINE | ID: mdl-31306644

ABSTRACT

Identifying novel therapeutics for the treatment of substance use disorder (SUD) is an area of intensive investigation. Prior strategies that have attempted to modify one or a few neurotransmitter receptors have had limited success, and currently there are no FDA-approved medications for the treatment of cocaine, methamphetamine, and marijuana use disorders. Because drugs of abuse are known to alter the expression of numerous genes in reward-related brain regions, epigenetic-based therapies have emerged as intriguing targets for therapeutic innovation. Here, I evaluate potential therapeutic approaches and challenges in targeting epigenetic factors for the treatment of SUD and highlight examples of promising strategies and future directions.


Subject(s)
Behavior, Addictive/drug therapy , Behavior, Addictive/genetics , Brain/drug effects , Epigenesis, Genetic/drug effects , Substance-Related Disorders/drug therapy , Substance-Related Disorders/genetics , Animals , Behavior, Addictive/metabolism , Brain/metabolism , Epigenesis, Genetic/physiology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Narcotic Antagonists/pharmacology , Narcotic Antagonists/therapeutic use , Nicotinic Antagonists/pharmacology , Nicotinic Antagonists/therapeutic use , Substance-Related Disorders/metabolism
8.
J Neurosci ; 39(4): 612-626, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30504275

ABSTRACT

Histone deacetylase (HDAC) inhibitors may have therapeutic utility in multiple neurological and psychiatric disorders, but the underlying mechanisms remain unclear. Here, we identify BRD4, a BET bromodomain reader of acetyl-lysine histones, as an essential component involved in potentiated expression of brain-derived neurotrophic factor (BDNF) and memory following HDAC inhibition. In in vitro studies, we reveal that pharmacological inhibition of BRD4 reversed the increase in BDNF mRNA induced by the class I/IIb HDAC inhibitor suberoylanilide hydroxamic acid (SAHA). Knock-down of HDAC2 and HDAC3, but not other HDACs, increased BDNF mRNA expression, whereas knock-down of BRD4 blocked these effects. Using dCas9-BRD4, locus-specific targeting of BRD4 to the BDNF promoter increased BDNF mRNA. In additional studies, RGFP966, a pharmacological inhibitor of HDAC3, elevated BDNF expression and BRD4 binding to the BDNF promoter, effects that were abrogated by JQ1 (an inhibitor of BRD4). Examining known epigenetic targets of BRD4 and HDAC3, we show that H4K5ac and H4K8ac modifications and H4K5ac enrichment at the BDNF promoter were elevated following RGFP966 treatment. In electrophysiological studies, JQ1 reversed RGFP966-induced enhancement of LTP in hippocampal slice preparations. Last, in behavioral studies, RGFP966 increased subthreshold novel object recognition memory and cocaine place preference in male C57BL/6 mice, effects that were reversed by cotreatment with JQ1. Together, these data reveal that BRD4 plays a key role in HDAC3 inhibitor-induced potentiation of BDNF expression, neuroplasticity, and memory.SIGNIFICANCE STATEMENT Some histone deacetylase (HDAC) inhibitors are known to have neuroprotective and cognition-enhancing properties, but the underlying mechanisms have yet to be fully elucidated. In the current study, we reveal that BRD4, an epigenetic reader of histone acetylation marks, is necessary for enhancing brain-derived neurotrophic factor (BDNF) expression and improved memory following HDAC inhibition. Therefore, by identifying novel epigenetic regulators of BDNF expression, these data may lead to new therapeutic targets for the treatment of neuropsychiatric disorders.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Histone Deacetylase Inhibitors/pharmacology , Memory/drug effects , Acrylamides/pharmacology , Animals , Azepines/pharmacology , Brain-Derived Neurotrophic Factor/drug effects , Epigenesis, Genetic , Gene Knockdown Techniques , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Phenylenediamines/pharmacology , Rats , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Triazoles/pharmacology , Vorinostat/pharmacology
9.
Proc Natl Acad Sci U S A ; 115(47): E11148-E11157, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30397132

ABSTRACT

Alzheimer's disease (AD) is the leading cause of age-related dementia. Neuropathological hallmarks of AD include brain deposition of ß-amyloid (Aß) plaques and accumulation of both hyperphosphorylated and acetylated tau. RGFP-966, a brain-penetrant and selective HDAC3 inhibitor, or HDAC3 silencing, increases BDNF expression, increases histone H3 and H4 acetylation, decreases tau phosphorylation and tau acetylation at disease-associated sites, reduces ß-secretase cleavage of the amyloid precursor protein (APP), and decreases Aß1-42 accumulation in HEK-293 cells overexpressing APP with the double Swedish mutation (HEK/APPsw). In the triple transgenic AD mouse model (3xTg-AD), repeated administration of 3 and 10 mg/kg of RGFP-966 reverses pathological tau phosphorylation at Thr181, Ser202, and Ser396, increases levels of the Aß degrading enzyme Neprilysin in plasma, decreases Aß1-42 protein levels in the brain and periphery, and improves spatial learning and memory. Finally, we show that RGFP-966 decreases Aß1-42 accumulation and both tau acetylation and phosphorylation at disease residues in neurons derived from induced pluripotent stem cells obtained from APOEε4-carrying AD patients. These data indicate that HDAC3 plays an important regulatory role in the expression and regulation of proteins associated with AD pathophysiology, supporting the notion that HDAC3 may be a disease-modifying therapeutic target.


Subject(s)
Acrylamides/pharmacology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Memory/drug effects , Phenylenediamines/pharmacology , Spatial Learning/drug effects , tau Proteins/metabolism , Acetylation/drug effects , Alzheimer Disease/drug therapy , Animals , Brain-Derived Neurotrophic Factor/metabolism , Gene Silencing , HEK293 Cells , Histone Deacetylases/genetics , Histones/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Mice, Transgenic , Neprilysin/blood , Neurons/cytology , Phosphorylation/drug effects
10.
Hypertension ; 71(4): 752-760, 2018 04.
Article in English | MEDLINE | ID: mdl-29483230

ABSTRACT

Cocaine abuse increases the risk of cardiovascular mortality and morbidity; however, the underlying molecular mechanisms remain elusive. By using a mouse model for cocaine abuse/use, we found that repeated cocaine injection led to increased blood pressure and aortic stiffness in mice associated with elevated levels of reactive oxygen species (ROS) in the aortas, a phenomenon similar to that observed in hypertensive humans. This ROS elevation was correlated with downregulation of Me1 (malic enzyme 1), an important redox molecule that counteracts ROS generation, and upregulation of microRNA (miR)-30c-5p that targets Me1 expression by directly binding to its 3'UTR (untranslated region). Remarkably, lentivirus-mediated overexpression of miR-30c-5p in aortic smooth muscle cells recapitulated the effect of cocaine on Me1 suppression, which in turn led to ROS elevation. Moreover, in vivo silencing of miR-30c-5p in smooth muscle cells resulted in Me1 upregulation, ROS reduction, and significantly suppressed cocaine-induced increases in blood pressure and aortic stiffness-a similar effect to that produced by treatment with the antioxidant N-acetyl cysteine. Discovery of this novel cocaine-↑miR-30c-5p-↓Me1-↑ROS pathway provides a potential new therapeutic avenue for treatment of cocaine abuse-related cardiovascular disease.


Subject(s)
Cocaine-Related Disorders , Cocaine/pharmacology , Malate Dehydrogenase/metabolism , MicroRNAs/metabolism , Reactive Oxygen Species/metabolism , Animals , Blood Pressure/drug effects , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/metabolism , Cocaine-Related Disorders/complications , Cocaine-Related Disorders/metabolism , Cocaine-Related Disorders/physiopathology , Disease Models, Animal , Down-Regulation , Injections , Mice , Oxidation-Reduction , Signal Transduction/drug effects , Signal Transduction/physiology , Vascular Stiffness/drug effects , Vasoconstrictor Agents/pharmacology
11.
Proc Natl Acad Sci U S A ; 114(43): E9135-E9144, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073110

ABSTRACT

Alzheimer's disease (AD) comprises multifactorial ailments for which current therapeutic strategies remain insufficient to broadly address the underlying pathophysiology. Epigenetic gene regulation relies upon multifactorial processes that regulate multiple gene and protein pathways, including those involved in AD. We therefore took an epigenetic approach where a single drug would simultaneously affect the expression of a number of defined AD-related targets. We show that the small-molecule histone deacetylase inhibitor M344 reduces beta-amyloid (Aß), reduces tau Ser396 phosphorylation, and decreases both ß-secretase (BACE) and APOEε4 gene expression. M344 increases the expression of AD-relevant genes: BDNF, α-secretase (ADAM10), MINT2, FE65, REST, SIRT1, BIN1, and ABCA7, among others. M344 increases sAPPα and CTFα APP metabolite production, both cleavage products of ADAM10, concordant with increased ADAM10 gene expression. M344 also increases levels of immature APP, supporting an effect on APP trafficking, concurrent with the observed increase in MINT2 and FE65, both shown to increase immature APP in the early secretory pathway. Chronic i.p. treatment of the triple transgenic (APPsw/PS1M146V/TauP301L) mice with M344, at doses as low as 3 mg/kg, significantly prevented cognitive decline evaluated by Y-maze spontaneous alternation, novel object recognition, and Barnes maze spatial memory tests. M344 displays short brain exposure, indicating that brief pulses of daily drug treatment may be sufficient for long-term efficacy. Together, these data show that M344 normalizes several disparate pathogenic pathways related to AD. M344 therefore serves as an example of how a multitargeting compound could be used to address the polygenic nature of multifactorial diseases.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Memory/drug effects , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Brain-Derived Neurotrophic Factor/genetics , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Maze Learning/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Memory/physiology , Mice, Transgenic , Peptide Fragments/metabolism , Repressor Proteins/genetics , Vorinostat
12.
Mol Cell Neurosci ; 85: 183-189, 2017 12.
Article in English | MEDLINE | ID: mdl-29055697

ABSTRACT

Natural antisense transcripts (NATs) are an abundant class of long noncoding RNAs that have recently been shown to be key regulators of chromatin dynamics and gene expression in nervous system development and neurological disorders. However, it is currently unclear if NAT-based mechanisms also play a role in drug-induced neuroadaptations. Aberrant regulation of gene expression is one critical factor underlying the long-lasting behavioral abnormalities that characterize substance use disorder, and it is possible that some drug-induced transcriptional responses are mediated, in part, by perturbations in NAT activity. To test this hypothesis, we used an automated algorithm that mines the NCBI AceView transcriptomics database to identify NAT overlapping genes linked to addiction. We found that 22% of the genes examined contain NATs and that expression of Homer1 natural antisense transcript (Homer1-AS) was altered in the nucleus accumbens (NAc) of mice 2h and 10days following repeated cocaine administration. In in vitro studies, depletion of Homer1-AS lead to an increase in the corresponding sense gene expression, indicating a potential regulatory mechanisms of Homer1 expression by its corresponding antisense transcript. Future in vivo studies are needed to definitely determine a role for Homer1-AS in cocaine-induced behavioral and molecular adaptations.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Homer Scaffolding Proteins/drug effects , Nucleus Accumbens/drug effects , RNA, Antisense/biosynthesis , Animals , Gene Expression Regulation/genetics , Homer Scaffolding Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Antisense/drug effects
13.
Curr Alzheimer Res ; 13(9): 985-95, 2016.
Article in English | MEDLINE | ID: mdl-27117003

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by welldefined neuropathological brain changes including amyloid plaques, neurofibrillary tangles and the presence of chronic neuroinflammation. OBJECTIVE: The brain penetrant BET bromodomain inhibitor JQ1 has been shown to regulate inflammation responses in vitro and in vivo, but its therapeutic potential in AD is currently unknown. METHOD: Three-month-old 3xTg mice were injected once a day with JQ1 (50 mg/kg) or vehicle for 15 weeks. At the end of the treatment learning and memory was assessed using the modified Barnes maze and the Y maze behavioral tests. Tissue from the brain and other organs was collected for molecular evaluation of neuroinflammation tau pathology and amyloid ß. RESULTS: JQ1 treatment reduced splenomegaly and neuroinflammation in the brain of treated mice where we observed a reduction in the expression of the pro-inflammatory modulators Il-1b, Il-6, Tnfa, Ccl2, Nos2 and Ptgs2. Additionally, JQ1-treated mice showed a reduction of tau phosphorylation at Ser396 in the hippocampus and frontal cortex while total levels of tau remained unaffected. On the other hand, JQ1 did not ameliorate learning and memory deficits in 7-month-old 3xTg mice. CONCLUSION: Taken together, our data suggest that BET bromodomain inhibitors hold the promise to be used for the treatment of neurological disorders characterized by neuroinflammation.


Subject(s)
Alzheimer Disease/drug therapy , Azepines/pharmacology , Brain/drug effects , Inflammation/drug therapy , Neuroprotective Agents/pharmacology , Triazoles/pharmacology , tau Proteins/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Brain/immunology , Brain/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Inflammation/metabolism , Inflammation/pathology , Learning/drug effects , Male , Memory/drug effects , Mice, Transgenic , Organ Size , Peptide Fragments/metabolism , Phosphorylation/drug effects , Spleen/drug effects , Spleen/immunology , Spleen/pathology
14.
J Neurosci ; 35(45): 15062-72, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26558777

ABSTRACT

Epigenetic processes that regulate histone acetylation play an essential role in behavioral and molecular responses to cocaine. To date, however, only a small fraction of the mechanisms involved in the addiction-associated acetylome have been investigated. Members of the bromodomain and extraterminal (BET) family of epigenetic "reader" proteins (BRD2, BRD3, BRD4, and BRDT) bind acetylated histones and serve as a scaffold for the recruitment of macromolecular complexes to modify chromatin accessibility and transcriptional activity. The role of BET proteins in cocaine-induced plasticity, however, remains elusive. Here, we used behavioral, pharmacological, and molecular techniques to examine the involvement of BET bromodomains in cocaine reward. Of the BET proteins, BRD4, but not BRD2 or BRD3, was significantly elevated in the nucleus accumbens (NAc) of mice and rats following repeated cocaine injections and self-administration. Systemic and intra-accumbal inhibition of BRD4 with the BET inhibitor, JQ1, attenuated the rewarding effects of cocaine in a conditioned place preference procedure but did not affect conditioned place aversion, nor did JQ1 alone induce conditioned aversion or preference. Investigating the underlying mechanisms, we found that repeated cocaine injections enhanced the binding of BRD4, but not BRD3, to the promoter region of Bdnf in the NAc, whereas systemic injection of JQ1 attenuated cocaine-induced expression of Bdnf in the NAc. JQ1 and siRNA-mediated knockdown of BRD4 in vitro also reduced expression of Bdnf. These findings indicate that disrupting the interaction between BET proteins and their acetylated lysine substrates may provide a new therapeutic avenue for the treatment of drug addiction. SIGNIFICANCE STATEMENT: Proteins involved in the "readout" of lysine acetylation marks, referred to as BET bromodomain proteins (including BRD2, BRD3, BRD4, and BRDT), have been shown to be key regulators of chromatin dynamics and disease, and BET inhibitors are currently being studied in several clinical trials. However, their role in addiction-related phenomena remains unknown. In the current studies, we revealed that BRD4 is elevated in the nucleus accumbens and recruited to promoter regions of addiction-related genes following repeated cocaine administration, and that inhibition of BRD4 attenuates transcriptional and behavioral responses to cocaine. Together, these studies reveal that BET inhibitors may have therapeutic utility in the treatment of cocaine addiction.


Subject(s)
Cocaine/pharmacology , Epigenesis, Genetic/physiology , Lysine/metabolism , Neuronal Plasticity/physiology , Acetylation/drug effects , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Epigenesis, Genetic/drug effects , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Rats , Rats, Sprague-Dawley
15.
Neuropsychopharmacology ; 39(5): 1059-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24257156

ABSTRACT

Recent studies have shown that post-retrieval extinction training attenuates fear and reward-related memories in both humans and rodents. This noninvasive, behavioral approach has the potential to be used in clinical settings to treat maladaptive memories that underlie several psychiatric disorders, including drug addiction. However, few studies to date have used a post-retrieval extinction approach to attenuate addiction-related memories. In the current study, we attempted to disrupt cocaine-related memories by using the post-retrieval extinction paradigm in male Sprague Dawley rats. Results revealed that starting extinction training 1 h after cocaine contextual memory was retrieved significantly attenuated cocaine-primed reinstatement of conditioned place preference (CPP) and relapse of cocaine CPP (drug-free and cocaine-primed) following 30 days of abstinence. However, animals that did not retrieve the contextual cocaine memory before extinction training, or animals that began extinction training 24 h after retrieval (outside of the reconsolidation window), demonstrated normal cocaine CPP. Conversely, animals that received additional CPP conditioning, rather than extinction training, 1 h after reactivation of cocaine memory showed enhanced cocaine CPP compared with animals that did not reactivate the cocaine memory before conditioning. These results reveal that a behavioral manipulation that takes advantage of reconsolidation and extinction of drug memories may be useful in decreasing preference for, and abuse of, cocaine.


Subject(s)
Cocaine-Related Disorders/therapy , Extinction, Psychological , Memory , Animals , Behavior Therapy , Behavior, Addictive/therapy , Cocaine/pharmacology , Conditioning, Psychological/drug effects , Dopamine Uptake Inhibitors/pharmacology , Extinction, Psychological/drug effects , Male , Memory/drug effects , Rats , Rats, Sprague-Dawley , Recurrence , Space Perception/drug effects , Task Performance and Analysis
16.
Eur J Neurosci ; 36(11): 3549-58, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23039920

ABSTRACT

Lateral hypothalamus (LH) orexin neurons are essential for the expression of a cocaine place preference. However, the afferents that regulate the activity of these orexin neurons during reward behaviors are not completely understood. Using tract tracing combined with Fos staining, we examined LH afferents for Fos induction during cocaine preference in rats. We found that the ventral bed nucleus of the stria terminalis (vBNST) was a major input to the LH orexin cell field that was significantly Fos-activated during cocaine conditioned place preference (CPP). Inactivation of the vBNST with baclofen plus muscimol blocked expression of cocaine CPP. Surprisingly, such inactivation of the vBNST also increased Fos induction in LH orexin neurons; as activity in these cells is normally associated with increased preference, this result indicates that a vBNST-orexin connection is unlikely to be responsible for CPP that is dependent on vBNST activity. Because previous studies have revealed that vBNST regulates dopamine cells in the ventral tegmental area (VTA), which is known to be involved in CPP and other reward functions, we tested whether vBNST afferents to the VTA are necessary for cocaine CPP. We found that disconnection of the vBNST and VTA (using local microinjections of baclofen plus muscimol unilaterally into the vBNST and contralateral VTA) significantly attenuated expression of cocaine preference. However, blocking ionotropic glutamatergic afferents to the VTA from the vBNST did not significantly reduce cocaine preference. These results indicate that a non-glutamatergic vBNST-VTA projection is involved in expression of cocaine preference.


Subject(s)
Cocaine/pharmacology , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/pharmacology , Septal Nuclei/physiology , Ventral Tegmental Area/physiology , Animals , Baclofen/pharmacology , Dopaminergic Neurons/physiology , Excitatory Amino Acid Antagonists/pharmacology , GABA-B Receptor Agonists/pharmacology , Male , Muscimol/pharmacology , Neuronal Tract-Tracers/analysis , Neurons, Afferent/physiology , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Sprague-Dawley
17.
Prog Brain Res ; 198: 79-121, 2012.
Article in English | MEDLINE | ID: mdl-22813971

ABSTRACT

Orexins/hypocretins are hypothalamic peptides involved in arousal and wakefulness, but also play a critical role in drug addiction and reward-related behaviors. Here, we review the roles played by orexins in a variety of animal models of drug addiction, emphasizing both commonalities and differences for orexin's involvement in seeking of the major classes of abused drugs, as well as food. One common theme that emerges is an involvement of orexins in drug seeking triggered by external stimuli (e.g., cues, contexts or stressors). We also discuss the functional neuronal circuits in which orexins are embedded, and how these circuits mediate addiction-related behaviors, with particular focus on the role of orexin and glutamate interactions within the ventral tegmental area. Finally, we attempt to contextualize the role of orexins in reward by discussing ways in which these peptides, expressed in only a few thousand neurons in the brain, can have such wide-ranging effects on behavior.


Subject(s)
Brain/physiopathology , Intracellular Signaling Peptides and Proteins/physiology , Motivation/physiology , Neuropeptides/physiology , Substance-Related Disorders/physiopathology , Animals , Humans , Orexins
18.
Front Genet ; 3: 106, 2012.
Article in English | MEDLINE | ID: mdl-22737160

ABSTRACT

Prolonged drug use causes long-lasting neuroadaptations in reward-related brain areas that contribute to addiction. Despite significant amount of research dedicated to understanding the underlying mechanisms of addiction, the molecular underpinnings remain unclear. At the same time, much of the pervasive transcription that encompasses the human genome occurs in the nervous system and contributes to its heterogeneity and complexity. Recent evidence suggests that non-coding RNAs (ncRNAs) play an important and dynamic role in transcriptional regulation, epigenetic signaling, stress response, and plasticity in the nervous system. Dysregulation of ncRNAs are thought to contribute to many, and perhaps all, neurological disorders, including addiction. Here, we review recent insights in the functional relevance of ncRNAs, including both microRNAs (miRNAs), and long non-coding RNAs, and then illustrate specific examples of ncRNA regulation in the context of drug addiction. We conclude that ncRNAs are importantly involved in the persistent neuroadaptations associated with addiction-related behaviors, and that therapies that target specific ncRNAs may represent new avenues for the treatment of drug addiction.

19.
J Neurosci ; 32(13): 4623-31, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22457508

ABSTRACT

Orexins (also called hypocretins) have been shown to be importantly involved in reward and addiction, but little is known about the circuitry that regulates orexin neuronal activity during drug-seeking behaviors. Here, we examined inputs to the lateral hypothalamus (LH) orexin cell field from the lateral septum (LS) using tract-tracing and Fos immunohistochemistry after cocaine (10 mg/kg) conditioned place preference (CPP) in Sprague Dawley rats. We found that neurons in rostral LS (LSr) that project to LH are Fos-activated in proportion to cocaine CPP, and that inhibition of LSr neurons with local baclofen and muscimol microinjection (0.3/0.03 nmol) blocks expression of Fos in LH orexin cells and cocaine preference. In addition, using local inactivation in LS and orexin antisense morpholinos in LH, we found that LSr influences on LH orexin neurons are critical for the expression of cocaine preference. These results indicate that LSr activates LH orexin neurons during cocaine place preference, and that this circuit is essential for expression of cocaine place preference.


Subject(s)
Conditioning, Psychological/physiology , Drug-Seeking Behavior/physiology , Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/physiology , Septum of Brain/physiology , Animals , Baclofen/administration & dosage , Baclofen/pharmacology , Benzoxazoles/pharmacology , Cocaine/antagonists & inhibitors , Cocaine/pharmacology , Conditioning, Psychological/drug effects , Drug-Seeking Behavior/drug effects , Hypothalamus/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Male , Microinjections , Morpholinos/administration & dosage , Morpholinos/pharmacology , Muscimol/administration & dosage , Muscimol/pharmacology , Naphthyridines , Neural Pathways/physiology , Neuroanatomical Tract-Tracing Techniques/methods , Neurons/physiology , Neuropeptides/antagonists & inhibitors , Orexins , Rats , Rats, Sprague-Dawley , Septum of Brain/drug effects , Urea/analogs & derivatives , Urea/pharmacology
20.
J Neurosci Methods ; 203(2): 354-60, 2012 Jan 30.
Article in English | MEDLINE | ID: mdl-22027492

ABSTRACT

Vivo-morpholinos are commercially available morpholino oligomers with a terminal octa-guanidinium dendrimer for enhanced cell-permeability. Existing evidence from systemically delivered vivo-morpholinos indicate that genetic suppression can last from days to weeks without evidence of cellular toxicity. However, intravenously delivered vivo-morpholinos are ineffective at protein suppression in the brain, and no evidence is available regarding whether intracranially delivered vivo-morpholinos effectively reduce target protein levels, or do so without inducing neurotoxicity. Here we report examples in which in vivo microinjection of antisense vivo-morpholinos directed against three different targets (xCT, GLT1, orexin) in two different brain regions resulted in significant suppression of protein expression without neurotoxicity. Expression was significantly suppressed at six to seven days post-administration, but returned to baseline levels within fourteen days. These results indicate that direct intracranial administration of vivo-morpholinos provides an effective means by which to suppress protein expression in the brain for one to two weeks.


Subject(s)
Antisense Elements (Genetics)/administration & dosage , Brain/metabolism , Morpholinos/administration & dosage , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Proteomics/methods , Animals , Antisense Elements (Genetics)/chemistry , Antisense Elements (Genetics)/genetics , Brain/cytology , Brain/drug effects , Male , Microinjections/methods , Morpholinos/chemistry , Morpholinos/genetics , Nerve Tissue Proteins/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...