Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Handb Exp Pharmacol ; 258: 373-393, 2020.
Article in English | MEDLINE | ID: mdl-31267166

ABSTRACT

Tobacco dependence is a leading cause of preventable disease and death worldwide. Nicotine, the main psychoactive component in tobacco cigarettes, has also been garnering increased popularity in its vaporized form, as derived from e-cigarette devices. Thus, an understanding of the molecular mechanisms underlying nicotine pharmacology and dependence is required to ascertain novel approaches to treat drug dependence. In this chapter, we review the field's current understanding of nicotine's actions in the brain, the neurocircuitry underlying drug dependence, factors that modulate the function of nicotinic acetylcholine receptors, and the role of specific genes in mitigating the vulnerability to develop nicotine dependence. In addition to nicotine's direct actions in the brain, other constituents in nicotine and tobacco products have also been found to alter drug use, and thus, evidence is provided to highlight this issue. Finally, currently available pharmacotherapeutic strategies are discussed, along with an outlook for future therapeutic directions to achieve to the goal of long-term nicotine cessation.


Subject(s)
Nicotine/pharmacology , Receptors, Nicotinic/physiology , Tobacco Use Disorder/physiopathology , Brain/drug effects , Humans
2.
J Neurosci ; 38(21): 4846-4858, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29712779

ABSTRACT

Dopamine is critical for processing of reward and etiology of drug addiction. Astrocytes throughout the brain express dopamine receptors, but consequences of astrocytic dopamine receptor signaling are not well established. We found that extracellular dopamine triggered rapid concentration-dependent stellation of astrocytic processes that was not a result of dopamine oxidation but instead relied on both cAMP-dependent and cAMP-independent dopamine receptor signaling. This was accompanied by reduced duration and increased frequency of astrocytic Ca2+ transients, but little effect on astrocytic voltage-gated potassium channel currents. To isolate possible mechanisms underlying these structural and functional changes, we used whole-genome RNA sequencing and found prominent dopamine-induced enrichment of genes containing the CCCTC-binding factor (CTCF) motif, suggesting involvement of chromatin restructuring in the nucleus. CTCF binding to promoter sites bidirectionally regulates gene transcription and depends on activation of poly-ADP-ribose polymerase 1 (PARP1). Accordingly, antagonism of PARP1 occluded dopamine-induced changes, whereas a PARP1 agonist facilitated dopamine-induced changes on its own. These results indicate that astrocyte response to elevated dopamine involves PARP1-mediated CTCF genomic restructuring and concerted expression of gene networks. Our findings propose epigenetic regulation of chromatin landscape as a critical factor in the rapid astrocyte response to dopamine.SIGNIFICANCE STATEMENT Although dopamine is widely recognized for its role in modulating neuronal responses both in healthy and disease states, little is known about dopamine effects at non-neuronal cells in the brain. To address this gap, we performed whole-genome sequencing of astrocytes exposed to elevated extracellular dopamine and combined it with evaluation of effects on astrocyte morphology and function. We demonstrate a temporally dynamic pattern of genomic plasticity that triggers pronounced changes in astrocyte morphology and function. We further show that this plasticity depends on activation of genes sensitive to DNA-binding protein CTCF. Our results propose that a broad pattern of astrocyte responses to dopamine specifically relies on CTCF-dependent gene networks.


Subject(s)
Astrocytes/drug effects , Astrocytes/ultrastructure , CCCTC-Binding Factor/drug effects , CCCTC-Binding Factor/genetics , Dopamine/pharmacology , Animals , CCCTC-Binding Factor/physiology , Calcium Signaling/drug effects , Chromatin/genetics , Chromatin/physiology , Electrophysiological Phenomena/physiology , Gene Expression Regulation/drug effects , Genomics , Poly (ADP-Ribose) Polymerase-1/drug effects , Poly (ADP-Ribose) Polymerase-1/genetics , Potassium Channels, Voltage-Gated/drug effects , RNA/genetics , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA , Transcriptome/drug effects , Transcriptome/genetics
3.
Int J Neuropsychopharmacol ; 19(1)2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26232787

ABSTRACT

BACKGROUND: Environmental enrichment alters susceptibility in developing drug addiction. We have demonstrated that rats raised in an enriched condition are more sensitive than rats raised in an impoverished condition to nicotine-induced locomotor activity, and this is associated with alterations of phosphorylated extracellular signal-regulated kinase 1/2 within the prefrontal cortex. This study determined the impact of microRNA-221 in the prefrontal cortex on phosphorylated extracellular signal-regulated kinase 1/2 and the enriched environment-dependent behavioral changes in response to nicotine. METHODS: A microRNA array was conducted to profile microRNA expression in the prefrontal cortex of enriched condition and impoverished condition rats in response to repeated nicotine (0.35 mg/kg, s.c.) administration. microRNA-221 in the prefrontal cortex, nucleus accumbens, and striatum was further verified by quantitative real-time PCR. Lentiviral-mediated overexpression of microRNA-221 in PC12 cells and the medial prefrontal cortex was performed to determine the effects of microRNA-221 on nicotine-mediated phosphorylated extracellular signal-regulated kinase 1/2, phosphorylated cAMP-response element-binding protein, and locomotor activity. RESULTS: microRNA-221 was profoundly upregulated in the prefrontal cortex but not in nucleus accumbens and striatum of enriched condition rats relative to impoverished condition rats following repeated administration of nicotine. Overexpression of lentiviral-microRNA-221 attenuated nicotine-induced increase in phosphorylated extracellular signal-regulated kinase 1/2 in PC12 cells. Lentiviral-microRNA-221 overexpression in the medial prefrontal cortex further increased locomotor activity in impoverished condition but not in enriched condition rats in response to repeated nicotine administration. Accordingly, lentiviral-microRNA-221 attenuated nicotine-induced increases in phosphorylated extracellular signal-regulated kinase 1/2 and phosphorylated cAMP-response element-binding protein in the medial prefrontal cortex of impoverished condition but not enriched condition rats. CONCLUSION: These findings suggest that environmental enrichment, via upregulation of prefrontal microRNA-221 expression, suppresses the nicotine-induced activation of extracellular signal-regulated kinase and cAMP-response element-binding protein, which provides a potential mechanism underlying enhanced locomotor sensitivity to nicotine.


Subject(s)
Environment , Locomotion/drug effects , MicroRNAs/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Prefrontal Cortex/drug effects , Animals , Animals, Newborn , CREB-Binding Protein/metabolism , Computational Biology , Gene Expression Profiling , HEK293 Cells , Humans , MAP Kinase Signaling System/drug effects , Male , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , PC12 Cells , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
4.
J Neurosci ; 33(22): 9451-61, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23719812

ABSTRACT

We previously showed that after repeated exposure to cocaine, D1-like dopamine receptor (D1DR) stimulation reverses plastic changes of AMPA receptor-mediated signaling in the nucleus accumbens shell. However, there is little information on the impact of cocaine self-administration on D1-NMDA receptor interactions in this brain region. Here, using whole-cell patch-clamp recordings, we assessed whether cocaine self-administration alters the effects of D1DR stimulation on synaptic and extrasynaptic NMDA receptors (NMDARs). In slices from cocaine-naive rats, pretreatment with a D1DR agonist decreased synaptic NMDAR-mediated currents and increased the contribution of extrasynaptic NMDARs. In contrast, neither cocaine self-administration alone nor cocaine experience followed by D1DR stimulation had an effect on synaptic or extrasynaptic NMDAR signaling. Activation of extrasynaptic NMDARs relies on the availability of extracellular glutamate, which is regulated primarily by glutamate transporters. In cocaine-experienced animals, relative to cocaine-naive rats, administration of a glutamate reuptake blocker, DL-threo-ß-benzyloxyaspartic acid, revealed increased extrasynaptic NMDAR activity and stronger baseline activity of glutamate uptake transporters. In cocaine-naive rats, the D1DR-mediated increase in extrasynaptic NMDAR signaling was independent of the activity of glutamate reuptake transporters. Together, these results indicate that cocaine experience blunts the influence of D1DRs on synaptic and extrasynaptic NMDAR signaling. Additionally, prior cocaine self-administration limits activation of the extrasynaptic NMDAR pool by increasing glutamate reuptake. These findings outline a pattern of adaptive interactions between D1DRs and NMDARs in the nucleus accumbens shell and demonstrate upregulation of extrasynaptic NMDAR signaling as a novel consequence of cocaine self-administration.


Subject(s)
Cocaine-Related Disorders/psychology , Cocaine/pharmacology , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Receptors, Dopamine D1/agonists , Receptors, N-Methyl-D-Aspartate/physiology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Amino Acid Transport System X-AG/antagonists & inhibitors , Amino Acid Transport System X-AG/metabolism , Animals , Aspartic Acid/pharmacology , Blotting, Western , Data Interpretation, Statistical , Extracellular Space/physiology , Glutamic Acid/physiology , Male , Nucleus Accumbens/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Self Administration , Synapses/physiology , Up-Regulation
5.
J Neurochem ; 129(4): 721-31, 2014 May.
Article in English | MEDLINE | ID: mdl-24422997

ABSTRACT

Chronic nicotine administration increases the density of brain α4ß2* nicotinic acetylcholine receptors (nAChRs), which may contribute to nicotine addiction by exacerbating withdrawal symptoms associated with smoking cessation. Varenicline, a smoking cessation drug, also increases these receptors in rodent brain. The maintenance of this increase by varenicline as well as nicotine replacement may contribute to the high rate of relapse during the first year after smoking cessation. Recently, we found that sazetidine-A (saz-A), a potent partial agonist that desensitizes α4ß2* nAChRs, does not increase the density of these receptors in brain at doses that decrease nicotine self-administration, increase attention in rats, and produce anxiolytic effects in mice. Here, we investigated whether chronic saz-A and varenicline maintain the density of nAChRs after their up-regulation by nicotine. In addition, we examined the effects of these drugs on a measure of anxiety in mice and weight gain in rats. After increasing nAChRs in the rodent brain with chronic nicotine, replacing nicotine with chronic varenicline maintained the increased nAChR binding, as well as the α4ß2 subunit proteins measured by western blots. In contrast, replacing nicotine treatments with chronic saz-A resulted in the return of the density of nAChRs to the levels seen in saline controls. Nicotine, saz-A and varenicline each demonstrated anxiolytic effects in mice, but only saz-A and nicotine attenuated the gain of weight over a 6-week period in rats. These findings suggest that apart from its modest anxiolytic and weight control effects, saz-A, or drugs like it, may be useful in achieving long-term abstinence from smoking.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety/prevention & control , Azetidines/therapeutic use , Brain Chemistry/drug effects , Nicotine/toxicity , Nicotinic Agonists/therapeutic use , Pyridines/therapeutic use , Receptors, Nicotinic/biosynthesis , Substance Withdrawal Syndrome/prevention & control , Tobacco Use Disorder/drug therapy , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacology , Anxiety/chemically induced , Azetidines/administration & dosage , Azetidines/pharmacology , Benzazepines/administration & dosage , Benzazepines/pharmacology , Benzazepines/therapeutic use , Drug Evaluation, Preclinical , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Quinoxalines/administration & dosage , Quinoxalines/pharmacology , Quinoxalines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/genetics , Tobacco Use Cessation , Tobacco Use Disorder/metabolism , Up-Regulation/drug effects , Varenicline , Weight Gain/drug effects
6.
J Pharmacol Exp Ther ; 349(2): 348-54, 2014 May.
Article in English | MEDLINE | ID: mdl-24627467

ABSTRACT

Although nicotine mediates its effects through several nicotinic acetylcholine receptor (nAChR) subtypes, it remains to be determined which nAChR subtypes directly mediate heightened anxiety during withdrawal. Relative success in abstinence has been found with the nAChR partial agonist varenicline (Chantix; Pfizer, Groton, CT); however, treatment with this drug fails to alleviate anxiety in individuals during nicotine withdrawal. Therefore, it is hypothesized that success can be found by the repurposing of other nAChR partial agonists for cessation therapies that target anxiety. It is noteworthy that the selective partial agonists for α4ß2, ABT-089 [2-methyl-3-[2(S)-pyrrolidinylmethoxy]pyridine], and α7, ABT-107 [5-(6-[(3R)-1-azabicyclo[2.2.2]oct-3-yloxy] pyridazin-3-yl)-1H-indole] (AbbVie, North Chicago, IL), have not been evaluated as possible therapeutics for nicotine cessation. Therefore, we examined the effect of ABT-089 and ABT-107 on anxiety during withdrawal from nicotine in the novelty-induced hypophagia (NIH) paradigm. We found that short-term administration of ABT-089 and ABT-107 alleviate anxiety-like behavior during withdrawal from nicotine while long-term administration of ABT-089 but not ABT-107 reduces anxiety-like behavior during withdrawal. After behavioral testing, brains were harvested and ß2-containing nAChRs were measured using [(3)H]epibaditine. ABT-089 and ABT-107 do not upregulate nAChRs, which is in contrast to the upregulation of nAChRs observed after nicotine. Furthermore, ABT-089 is anxiogenic in nicotine naive animals, suggesting that the effects on anxiety are specifically related to the nicotine-dependent state. Together, these studies identify additional nAChR partial agonists that may aid in the rational development of smoking cessation aids.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Indoles/pharmacology , Nicotine/adverse effects , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Pyrrolidines/pharmacology , Quinuclidines/pharmacology , Receptors, Nicotinic/metabolism , Substance Withdrawal Syndrome/drug therapy , Animals , Anti-Anxiety Agents/therapeutic use , Anxiety/metabolism , Anxiety/psychology , Brain/metabolism , Drug Partial Agonism , Indoles/therapeutic use , Male , Mice , Nicotinic Agonists/therapeutic use , Pyridines/therapeutic use , Pyrrolidines/therapeutic use , Quinuclidines/therapeutic use , Radioligand Assay , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology , Time Factors , Up-Regulation
7.
Adv Pharmacol ; 99: 387-404, 2024.
Article in English | MEDLINE | ID: mdl-38467488

ABSTRACT

Nicotine use disorder remains a major public health emergency despite years of trumpeting the consequences of smoking. This is likely due to the complex interplay of genetics and nicotine exposure across the lifespan of these individuals. Genetics influence all aspects of life, including complex disorders such as nicotine use disorder. This review first highlights the critical neurocircuitry underlying nicotine dependence and withdrawal, and then describes the cellular signaling mechanisms involved. Finally, current genetic, genomic, and transcriptomic evidence for new drug development of smoking cessation aids is discussed, with a focus on the Neuregulin 3 Signaling Pathway.


Subject(s)
Smoking Cessation , Tobacco Use Disorder , Humans , Tobacco Use Disorder/drug therapy , Tobacco Use Disorder/genetics , Tobacco Use Disorder/metabolism , Precision Medicine , Smoking/genetics , Neuregulins/genetics , Neuregulins/metabolism
8.
Biol Psychiatry Glob Open Sci ; 4(1): 182-193, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38298802

ABSTRACT

Background: Smoking is the largest preventable cause of death and disease in the United States, with <5% of quit attempts being successful. Microglia activation and proinflammatory neuroimmune signaling in reward neurocircuitry are implicated in nicotine withdrawal symptomology. Microglia are integral regulators of blood-brain barrier (BBB) functionality as well; however, whether the effects of nicotine withdrawal on microglia function impact BBB integrity is unknown. Methods: Mice were treated with chronic nicotine (12 mg/kg/day) and subjected to 48 hours nicotine withdrawal. Regional BBB permeability, together with messenger RNA and protein expression of tight junction proteins, were assessed. PLX5622 chow was used to deplete microglia to evaluate the role of microglia in regulating BBB integrity and nicotine withdrawal symptomology. Results: Female mice had higher baseline BBB permeability in the prefrontal cortex and hippocampus than males. Nicotine withdrawal further exacerbated the BBB permeability selectively in the prefrontal cortex of females. These effects were concurrent with prefrontal cortex alterations in a subset of tight junction proteins with increased proinflammatory responses following nicotine withdrawal in females. Depletion of microglia via PLX5622 treatment prevented all these molecular effects and attenuated withdrawal-induced anxiety-like behavior in female mice. Conclusions: These results are the first to show sex differences in regional BBB permeability during nicotine withdrawal. This represents a possible link to both the reduced smoking cessation success seen in women and women's increased risk for smoking-related neurovascular disorders. Furthermore, these findings open an avenue for sex-specific therapeutics that target microglia and BBB dysfunction during nicotine withdrawal in women.

9.
Neuropharmacology ; 247: 109846, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38211698

ABSTRACT

Tobacco smoking remains a leading cause of preventable death in the United States, with approximately a 5% success rate for smokers attempting to quit. High relapse rates have been linked to several genetic factors, indicating that the mechanistic relationship between genes and drugs of abuse is a valuable avenue for the development of novel smoking cessation therapies. For example, various single nucleotide polymorphisms (SNPs) in the gene for neuregulin 3 (NRG3) and its cognate receptor, the receptor tyrosine-protein kinase erbB-4 (ERBB4), have been linked to nicotine addiction. Our lab has previously shown that ERBB4 plays a role in anxiety-like behavior during nicotine withdrawal (WD); however, the neuronal mechanisms and circuit-specific effects of NRG3-ERBB4 signaling during nicotine and WD are unknown. The present study utilizes genetic, biochemical, and functional approaches to examine the anxiety-related behavioral and functional role of NRG3-ERBB4 signaling, specifically in the ventral hippocampus (VH) of male and female mice. We report that 24hWD from nicotine is associated with altered synaptic expression of VH NRG3 and ERBB4, and genetic disruption of VH ErbB4 leads to an elimination of anxiety-like behaviors induced during 24hWD. Moreover, we observed attenuation of GABAergic transmission as well as alterations in Ca2+-dependent network activity in the ventral CA1 area of VH ErbB4 knock-down mice during 24hWD. Our findings further highlight contributions of the NRG3-ERBB4 signaling pathway to anxiety-related behaviors seen during nicotine WD.


Subject(s)
Nicotine , Substance Withdrawal Syndrome , Male , Female , Mice , Animals , Nicotine/pharmacology , Nicotine/metabolism , Neuregulins/genetics , Neuregulins/metabolism , Substance Withdrawal Syndrome/metabolism , Hippocampus/metabolism , Signal Transduction , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism
10.
Neuropsychopharmacology ; 49(3): 551-560, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37660129

ABSTRACT

Dopaminergic signaling in the nucleus accumbens shell (NAc) regulates neuronal activity relevant to reward-related learning, including cocaine-associated behaviors. Although astrocytes respond to dopamine and cocaine with structural changes, the impact of dopamine and cocaine on astrocyte functional plasticity has not been widely studied. Specifically, behavioral implications of voltage-gated channel activity in the canonically non-excitable astrocytes are not known. We characterized potassium channel function in NAc astrocytes following exposure to exogenous dopamine or cocaine self-administration training under short (2 h/day) and extended (6 h/day) access schedules. Electrophysiological, Ca2+ imaging, mRNA, and mass spectrometry tools were used for molecular characterization. Behavioral effects were examined after NAc-targeted microinjections of channel antagonists and astroglial toxins. Exogenous dopamine increased activity of currents mediated by voltage-gated (Kv7) channels in NAc astrocytes. This was associated with a ~5-fold increase in expression of Kcnq2 transcript level in homogenized NAc micropunches. Matrix-assisted laser desorption/ionization mass spectrometry revealed increased NAc dopamine levels in extended access, relative to short access, rats. Kv7 inhibition selectively increased frequency and amplitude of astrocyte intracellular Ca2+ transients in NAc of extended access rats. Inhibition of Kv7 channels in the NAc attenuated cocaine-seeking in extended access rats only, an effect that was occluded by microinjection of the astrocyte metabolic poison, fluorocitrate. These results suggest that voltage-gated K+ channel signaling in NAc astrocytes is behaviorally relevant, support Kv7-mediated regulation of astrocyte Ca2+ signals, and propose novel mechanisms of neuroglial interactions relevant to drug use.


Subject(s)
Cocaine , Potassium Channels, Voltage-Gated , Rats , Animals , Astrocytes , Potassium Channels, Voltage-Gated/pharmacology , Rats, Sprague-Dawley , Dopamine/pharmacology , Nucleus Accumbens
11.
J Med Chem ; 67(11): 9173-9193, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38810170

ABSTRACT

While in the process of designing more effective synthetic opioid rescue agents, we serendipitously identified a new chemotype of potent synthetic opioid. Here, we report that conformational constraint of a piperazine ring converts a mu opioid receptor (MOR) antagonist into a potent MOR agonist. The prototype of the series, which we have termed atoxifent (2), possesses potent in vitro agonist activity. In mice, atoxifent displayed long-lasting antinociception that was reversible with naltrexone. Repeated dosing of atoxifent produced antinociceptive tolerance and a level of withdrawal like that of fentanyl. In rats, while atoxifent produced complete loss of locomotor activity like fentanyl, it failed to produce deep respiratory depression associated with fentanyl-induced lethality. Assessment of brain biodistribution demonstrated ample distribution of atoxifent into the brain with a Tmax of approximately 0.25 h. These results indicate enhanced safety for atoxifent-like molecules compared to fentanyl.


Subject(s)
Analgesics, Opioid , Fentanyl , Receptors, Opioid, mu , Respiratory Insufficiency , Animals , Mice , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/drug therapy , Analgesics, Opioid/pharmacology , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/chemistry , Rats , Male , Fentanyl/pharmacology , Fentanyl/chemical synthesis , Fentanyl/chemistry , Structure-Activity Relationship , Piperazines/pharmacology , Piperazines/chemistry , Piperazines/chemical synthesis , Piperazines/therapeutic use , Piperazines/pharmacokinetics , Humans , Rats, Sprague-Dawley , Tissue Distribution , Brain/metabolism , Brain/drug effects , Naltrexone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/chemical synthesis , Naltrexone/chemistry , Naltrexone/therapeutic use
12.
bioRxiv ; 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36711798

ABSTRACT

Tobacco smoking remains a leading cause of preventable death in the United States, with a less than 5% success rate for smokers attempting to quit. High relapse rates have been linked to several genetic factors, indicating that the mechanistic relationship between genes and drugs of abuse is a valuable avenue for the development of novel smoking cessation therapies. For example, various single nucleotide polymorphisms (SNPs) in the gene for neuregulin 3 (NRG3) and its cognate receptor, the receptor tyrosine-protein kinase erbB-4 (ERBB4), have been linked to nicotine addiction. Our lab has previously shown that ERBB4 plays a role in anxiety-like behavior during nicotine withdrawal (WD); however, the neuronal mechanisms and circuit-specific effects of NRG3-ERBB4 signaling during nicotine and WD are unknown. The present study utilizes genetic, biochemical, and functional approaches to examine the anxiety-related behavioral and functional role of NRG3-ERBB4 signaling, specifically in the ventral hippocampus (VH). We report that 24hWD from nicotine is associated with altered synaptic expression of VH NRG3 and ERBB4, and genetic disruption of VH ErbB4 leads to an elimination of anxiety-like behaviors induced during 24hWD. Moreover, we observed attenuation of GABAergic transmission as well as alterations in Ca2+-dependent network activity in the ventral CA1 area of VH ErbB4 knock-down mice during 24hWD. Our findings further highlight contributions of the NRG3-ERBB4 signaling pathway to anxiety-related behaviors seen during nicotine WD.

13.
J Pharmacol Exp Ther ; 343(2): 441-50, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22899752

ABSTRACT

Chronic nicotine administration increases α4ß2 neuronal nicotinic acetylcholine receptor (nAChR) density in brain. This up-regulation probably contributes to the development and/or maintenance of nicotine dependence. nAChR up-regulation is believed to be triggered at the ligand binding site, so it is not surprising that other nicotinic ligands also up-regulate nAChRs in the brain. These other ligands include varenicline, which is currently used for smoking cessation therapy. Sazetidine-A (saz-A) is a newer nicotinic ligand that binds with high affinity and selectivity at α4ß2* nAChRs. In behavioral studies, saz-A decreases nicotine self-administration and increases performance on tasks of attention. We report here that, unlike nicotine and varenicline, chronic administration of saz-A at behaviorally active and even higher doses does not up-regulate nAChRs in rodent brains. We used a newly developed method involving radioligand binding to measure the concentrations and nAChR occupancy of saz-A, nicotine, and varenicline in brains from chronically treated rats. Our results indicate that saz-A reached concentrations in the brain that were ∼150 times its affinity for α4ß2* nAChRs and occupied at least 75% of nAChRs. Thus, chronic administration of saz-A did not up-regulate nAChRs despite it reaching brain concentrations that are known to bind and desensitize virtually all α4ß2* nAChRs in brain. These findings reinforce a model of nicotine addiction based on desensitization of up-regulated nAChRs and introduce a potential new strategy for smoking cessation therapy in which drugs such as saz-A can promote smoking cessation without maintaining nAChR up-regulation, thereby potentially increasing the rate of long-term abstinence from nicotine.


Subject(s)
Azetidines/pharmacology , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Pyridines/pharmacology , Receptors, Nicotinic/drug effects , Animals , Azetidines/administration & dosage , Azetidines/blood , Benzazepines/administration & dosage , Benzazepines/blood , Benzazepines/pharmacology , Binding Sites , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Female , Male , Membranes/drug effects , Membranes/metabolism , Neurons/drug effects , Neurons/metabolism , Nicotine/administration & dosage , Nicotine/blood , Nicotine/pharmacology , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/blood , Nicotinic Agonists/pharmacology , Pregnancy , Pyridines/administration & dosage , Pyridines/blood , Quinoxalines/administration & dosage , Quinoxalines/blood , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Self Administration , Up-Regulation/drug effects , Varenicline
14.
Neurobiol Learn Mem ; 97(4): 482-94, 2012 May.
Article in English | MEDLINE | ID: mdl-22521799

ABSTRACT

Pre-adolescence and adolescence are developmental periods associated with increased vulnerability for tobacco addiction, and exposure to tobacco during these periods may lead to long-lasting changes in behavioral and neuronal plasticity. The present study examined the short- and long-term effects of nicotine and nicotine withdrawal on fear conditioning in pre-adolescent, adolescent, and adult mice, and potential underlying substrates that may mediate the developmental effects of nicotine, such as changes in nicotinic acetylcholine receptor (nAChR) binding, CREB expression, and nicotine metabolism. Age-related differences existed in sensitivity to the effects of acute nicotine, chronic nicotine and nicotine withdrawal on contextual fear conditioning (no changes in cued fear conditioning were seen); younger mice were more sensitive to the acute effects and less sensitive to the effects of nicotine withdrawal 24 h post treatment cessation. Developmental differences in nAChR binding were associated with the effects of nicotine withdrawal on contextual learning. Developmental differences in nicotine metabolism and CREB expression were also observed, but were not related to the effects of nicotine withdrawal on contextual learning 24 h post treatment. Chronic nicotine exposure during pre-adolescence or adolescence, however, produced long-lasting impairments in contextual learning that were observed during adulthood, whereas adult chronic nicotine exposure did not. These developmental effects could be related to changes in CREB. Overall, there is a developmental shift in the effects of nicotine on hippocampus-dependent learning and developmental exposure to nicotine results in adult cognitive deficits; these changes in cognition may play an important role in the development and maintenance of nicotine addiction.


Subject(s)
CREB-Binding Protein/metabolism , Conditioning, Psychological/drug effects , Hippocampus/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Substance Withdrawal Syndrome/physiopathology , Age Factors , Animals , CREB-Binding Protein/drug effects , Fear/drug effects , Male , Mice , Nicotine/adverse effects , Nicotine/metabolism , Nicotinic Agonists/adverse effects , Nicotinic Agonists/metabolism , Receptors, Nicotinic/drug effects , Tobacco Use Disorder , Up-Regulation/drug effects
15.
Neurosci Biobehav Rev ; 134: 104507, 2022 03.
Article in English | MEDLINE | ID: mdl-34968525

ABSTRACT

Concomitant use of tobacco and opioids represents a growing public health concern. In fact, the mortality rate due to smoking-related illness approaches 50% among SUD patients. Cumulative evidence demonstrates that the vulnerability to drugs of abuse is influenced by behavioral, environmental, and genetic factors. This review explores the contribution of genetics and neural mechanisms influencing nicotine and opioid reward, respiration, and antinociception, emphasizing the interaction of cholinergic and opioid receptor systems. Despite the substantial evidence demonstrating nicotine-opioid interactions within the brain and on behavior, the currently available pharmacotherapies targeting these systems have shown limited efficacy for smoking cessation on opioid-maintained smokers. Thus, further studies designed to identify novel targets modulating both nicotinic and opioid receptor systems may lead to more efficacious approaches for co-morbid nicotine dependence and opioid use disorder.


Subject(s)
Opioid-Related Disorders , Receptors, Nicotinic , Tobacco Use Disorder , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Humans , Nicotine/pharmacology , Nicotine/therapeutic use , Opioid-Related Disorders/drug therapy , Receptors, Nicotinic/therapeutic use , Tobacco Use Disorder/drug therapy
16.
Cerebellum ; 10(4): 748-57, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21562921

ABSTRACT

Although recent studies have delineated the specific nicotinic subtypes present in the mammalian cerebellum, very little is known about their location or function within the cerebellum. This is of increased interest since nicotinic receptors (nAChRs) in the cerebellum have recently been implicated in the pathology of autism spectrum disorders. To begin to better understand the roles of these heteromeric nAChRs in the cerebellar circuitry and their therapeutic potential as targets for drug development, we used various chemical and stereotaxic lesion models in conjunction with slice electrophysiology to examine how specific heteromeric nAChR subtypes may influence the surrounding cerebellar circuitry. Using subunit-specific immunoprecipitation of radiolabeled nAChRs in the cerebella following N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride, p-chloroamphetamine, and pendunculotomy lesions, we show that most, if not all, cerebellar nicotinic receptors are present in cells within the cerebellum itself and not in extracerebellar afferents. Furthermore, we demonstrate that the ß4-containing, but not the ß2-containing, nAChRs intrinsic to the cerebellum can regulate inhibitory synaptic efficacy at two major classes of cerebellar neurons. These tandem findings suggest that nAChRs may present a potential drug target for disorders involving the cerebellum.


Subject(s)
Cerebellum/metabolism , Receptors, Nicotinic/physiology , Animals , Cerebellum/drug effects , Cerebellum/physiology , Male , Nicotinic Agonists/pharmacology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
17.
Nicotine Tob Res ; 13(1): 41-6, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21097981

ABSTRACT

INTRODUCTION: Clinical and preclinical studies suggest that regulation of nicotinic acetylcholine receptors (nAChR) maybe involved in the etiology of withdrawal symptoms. METHODS: We evaluated heteromeric nAChR regulation via [³H]epibatidine binding following cessation of chronic nicotine or varenicline treatment. Animals were concurrently tested in the marble-burying test to evaluate treatment-related effects. RESULTS: We found that both nicotine (18 mg/kg/day, free base) and varenicline (1.8 mg/kg/day) chronically administered for 14 days upregulated nAChRs significantly in the cortex, hippocampus, striatum, and thalamus. The duration of upregulation (up to 72 hr) was both drug and region specific. In addition to nAChR upregulation, chronic administration of both nicotine and varenicline had anxiolytic-like effects in the marble-burying test. This effect was maintained for 48 hr following cessation of varenicline but was absent 24 hr following cessation from nicotine. Additionally, marble-burying behavior positively correlated to the regulation of cortical nAChRs following cessation of either treatment. CONCLUSIONS: Varenicline has been shown to be an efficacious smoking cessation aid, with a proposed mechanism of action that includes modulation of dopamine release in reward areas of the brain. Our studies show that varenicline elicits both anxiolytic effects in the marble-burying test as well as region- and time-specific receptor upregulation. These findings suggest receptor upregulation as a mechanism for its efficacy as a smoking cessation therapy.


Subject(s)
Benzazepines/pharmacology , Nicotine/pharmacology , Quinoxalines/pharmacology , Receptors, Nicotinic/metabolism , Up-Regulation/drug effects , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Smoking Cessation , Substance Withdrawal Syndrome/metabolism , Thalamus/drug effects , Thalamus/metabolism , Varenicline
18.
Article in English | MEDLINE | ID: mdl-32513669

ABSTRACT

While commendable strides have been made in reducing smoking initiation and improving smoking cessation rates, current available smoking cessation treatment options are still only mildly efficacious and show substantial interindividual variability in their therapeutic responses. Therefore, the primary goal of preclinical research has been to further the understanding of the neural substrates and genetic influences involved in nicotine's effects and reassess potential drug targets. Pronounced advances have been made by investing in new translational approaches and placing more emphasis on bridging the gap between human and rodent models of dependence. Functional neuroimaging studies have identified key brain structures involved with nicotine-dependence phenotypes such as craving, impulsivity, withdrawal symptoms, and smoking cessation outcomes. Following up with these findings, rodent-modeling techniques have made it possible to dissect the neural circuits involved in these motivated behaviors and ascertain mechanisms underlying nicotine's interactive effects on brain structure and function. Likewise, translational studies investigating single-nucleotide polymorphisms (SNPs) within the cholinergic, dopaminergic, and opioid systems have found high levels of involvement of these neurotransmitter systems in regulating the reinforcing aspects of nicotine in both humans and mouse models. These findings and coordinated efforts between human and rodent studies pave the way for future work determining gene by drug interactions and tailoring treatment options to each individual smoker.


Subject(s)
Tobacco Use Disorder/psychology , Translational Research, Biomedical , Animals , Humans , Models, Animal , Nicotine/administration & dosage , Rodentia
19.
Neuropharmacology ; 186: 108469, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33485944

ABSTRACT

Opioid use disorder is a leading cause of morbidity and mortality in the United States. Increasing pre-clinical and clinical evidence demonstrates sex differences in opioid use and dependence. However, the underlying molecular mechanisms contributing to these effects, including neuroinflammation, are still obscure. Therefore, in this study, we investigated the effect of oxycodone exposure and withdrawal on sex- and region-specific neuroimmune response. Real-time PCR and multiplex cytokine array analysis demonstrated elevated neuroinflammation with increased pro-inflammatory cytokine levels, and aberrant oligodendroglial response in reward neurocircuitry, following withdrawal from chronic oxycodone treatment. Chronic oxycodone and withdrawal treated male mice had lower mRNA expression of TMEM119 along with elevated protein levels of pro-inflammatory cytokines/chemokines and growth factors (IL-1ß, IL-2, IL-7, IL-9, IL-12, IL-15, IL17, M-CSF, VEGF) in the prefrontal cortex (PFC) as compared to their female counterparts. In contrast, reduced levels of pro-inflammatory cytokines/chemokines (IL-1ß, IL-6, IL-9, IL-12, CCL11) was observed in the nucleus accumbens (NAc) of oxycodone and withdrawal-treated males as compared to female mice. No treatment specific effects were observed on the mRNA expression of putative microglial activation markers (Iba1, CD68), but an overall sex specific decrease in the mRNA expression of Iba1 and CD68 was found in the PFC and NAc of male mice as compared to females. Moreover, a sex and region-specific increase in the mRNA levels of oligodendrocyte lineage markers (NG2, Sox10) was also observed in oxycodone and withdrawal treated animals. These findings may open a new avenue for the development of sex-specific precision therapeutics for opioid dependence by targeting region-specific neuroimmune signaling.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/toxicity , Neuroimmunomodulation/immunology , Sex Characteristics , Substance Withdrawal Syndrome/immunology , Substance Withdrawal Syndrome/metabolism , Animals , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Neuroimmunomodulation/drug effects , Oxycodone/administration & dosage , Oxycodone/toxicity , Prefrontal Cortex/drug effects , Prefrontal Cortex/immunology , Prefrontal Cortex/metabolism , Substance Withdrawal Syndrome/diagnosis
20.
J Pharmacol Exp Ther ; 334(2): 665-72, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20435920

ABSTRACT

Clinical and preclinical studies suggest that nicotinic acetylcholine receptors are involved in affective disorders; therefore, the potential therapeutic value of nicotinic partial agonists as treatments of these disorders is of growing interest. This study evaluated the effects of acute and chronic administration of nicotine and the alpha4beta2 nicotinic partial agonists varenicline and sazetidine-A in mouse models of anxiety and depression. Acutely, only nicotine and varenicline had anxiolytic effects in the marble-burying test and in the novelty-induced hypophagia (NIH) test. In contrast, in animal models of antidepressant efficacy, such as the forced swim and the tail suspension test, only acute sazetidine-A had significant antidepressant-like effects. The NIH test provides an anxiety-related measure that is sensitive to the effects of chronic but not acute antidepressant treatment. Chronic nicotine and chronic sazetidine-A treatment were effective in this paradigm, but varenicline was ineffective. These results suggest that the partial agonists varenicline and sazetidine-A may have diverse therapeutic benefits in affective disorders.


Subject(s)
Azetidines/pharmacology , Behavior, Animal/drug effects , Benzazepines/pharmacology , Mood Disorders/drug therapy , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Quinoxalines/pharmacology , Animals , Anxiety/drug therapy , Anxiety/psychology , Azetidines/therapeutic use , Benzazepines/therapeutic use , Chimera , Depression/drug therapy , Depression/psychology , Drug Partial Agonism , Male , Mice , Mood Disorders/psychology , Nicotine/pharmacology , Nicotinic Agonists/therapeutic use , Pyridines/therapeutic use , Quinoxalines/therapeutic use , Varenicline
SELECTION OF CITATIONS
SEARCH DETAIL