Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 42
1.
bioRxiv ; 2024 Feb 28.
Article En | MEDLINE | ID: mdl-38463990

Loss of dopamine neurons causes motor deterioration in Parkinson's disease patients. We have previously reported that in addition to acute motor impairment, the impaired motor behavior is encoded into long-term memory in an experience-dependent and task-specific manner, a phenomenon we refer to as aberrant inhibitory motor learning. Although normal motor learning and aberrant inhibitory learning oppose each other and this is manifested in apparent motor performance, in the present study, we found that normal motor memory acquired prior to aberrant inhibitory learning remains preserved in the brain, suggesting the existence of independent storage. To investigate the neuronal circuits underlying these two opposing memories, we took advantage of the RNA-binding protein YTHDF1, an m 6 A RNA methylation reader involved in the regulation of protein synthesis and learning/memory. Conditional deletion of Ythdf1 in either D1 or D2 receptor-expressing neurons revealed that normal motor memory is stored in the D1 (direct) pathway of the basal ganglia, while inhibitory memory is stored in the D2 (indirect) pathway. Furthermore, fiber photometry recordings of GCaMP signals from striatal D1 (dSPN) and D2 (iSPN) receptor-expressing neurons support the preservation of normal memory in the direct pathway after aberrant inhibitory learning, with activities of dSPN predictive of motor performance. Finally, a computational model based on activities of motor cortical neurons, dSPN and iSPN neurons, and their interactions through the basal ganglia loops supports the above observations. These findings have important implications for novel approaches in treating Parkinson's disease by reactivating preserved normal memory, and in treating hyperkinetic movement disorders such as chorea or tics by erasing aberrant motor memories.

2.
Neuron ; 111(21): 3414-3434.e15, 2023 11 01.
Article En | MEDLINE | ID: mdl-37734381

Chronic pain is a tremendous burden for afflicted individuals and society. Although opioids effectively relieve pain, significant adverse outcomes limit their utility and efficacy. To investigate alternate pain control mechanisms, we explored cholinergic signaling in the ventrolateral periaqueductal gray (vlPAG), a critical nexus for descending pain modulation. Biosensor assays revealed that pain states decreased acetylcholine release in vlPAG. Activation of cholinergic projections from the pedunculopontine tegmentum to vlPAG relieved pain, even in opioid-tolerant conditions, through ⍺7 nicotinic acetylcholine receptors (nAChRs). Activating ⍺7 nAChRs with agonists or stimulating endogenous acetylcholine inhibited vlPAG neuronal activity through Ca2+ and peroxisome proliferator-activated receptor α (PPAR⍺)-dependent signaling. In vivo 2-photon imaging revealed that chronic pain induces aberrant excitability of vlPAG neuronal ensembles and that ⍺7 nAChR-mediated inhibition of these cells relieves pain, even after opioid tolerance. Finally, pain relief through these cholinergic mechanisms was not associated with tolerance, reward, or withdrawal symptoms, highlighting its potential clinical relevance.


Chronic Pain , Receptors, Nicotinic , Rats , Animals , Humans , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Chronic Pain/drug therapy , Acetylcholine , Rats, Sprague-Dawley , Pain Measurement/methods , Drug Tolerance/physiology , Periaqueductal Gray/physiology , Cholinergic Agents/pharmacology , Receptors, Nicotinic/metabolism
3.
Nat Commun ; 9(1): 2710, 2018 07 13.
Article En | MEDLINE | ID: mdl-30006624

Nicotine use can lead to dependence through complex processes that are regulated by both its rewarding and aversive effects. Recent studies show that aversive nicotine doses activate excitatory inputs to the interpeduncular nucleus (IPN) from the medial habenula (MHb), but the downstream targets of the IPN that mediate aversion are unknown. Here we show that IPN projections to the laterodorsal tegmentum (LDTg) are GABAergic using optogenetics in tissue slices from mouse brain. Selective stimulation of these IPN axon terminals in LDTg in vivo elicits avoidance behavior, suggesting that these projections contribute to aversion. Nicotine modulates these synapses in a concentration-dependent manner, with strong enhancement only seen at higher concentrations that elicit aversive responses in behavioral tests. Optogenetic inhibition of the IPN-LDTg connection blocks nicotine conditioned place aversion, suggesting that the IPN-LDTg connection is a critical part of the circuitry that mediates the aversive effects of nicotine.


Avoidance Learning/physiology , GABAergic Neurons/drug effects , Habenula/drug effects , Interpeduncular Nucleus/drug effects , Nicotine/pharmacology , Tegmentum Mesencephali/drug effects , Animals , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Electrodes, Implanted , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gene Expression , Habenula/cytology , Habenula/metabolism , Interpeduncular Nucleus/cytology , Interpeduncular Nucleus/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Pathways/drug effects , Neural Pathways/metabolism , Optogenetics , Reward , Stereotaxic Techniques , Synapses/drug effects , Synapses/physiology , Tegmentum Mesencephali/cytology , Tegmentum Mesencephali/metabolism , Transgenes
4.
Pain ; 158(10): 1938-1950, 2017 Oct.
Article En | MEDLINE | ID: mdl-28817416

Along with the well-known rewarding effects, activation of nicotinic acetylcholine receptors (nAChRs) can also relieve pain, and some nicotinic agonists have analgesic efficacy similar to opioids. A major target of analgesic drugs is the descending pain modulatory pathway, including the ventrolateral periaqueductal gray (vlPAG) and the rostral ventromedial medulla (RVM). Although activating nAChRs within this circuitry can be analgesic, little is known about the subunit composition and cellular effects of these receptors, particularly within the vlPAG. Using electrophysiology in brain slices from adult male rats, we examined nAChR effects on vlPAG neurons that project to the RVM. We found that 63% of PAG-RVM projection neurons expressed functional nAChRs, which were exclusively of the α7-subtype. Interestingly, the neurons that express α7 nAChRs were largely nonoverlapping with those expressing µ-opioid receptors (MOR). As nAChRs are excitatory and MORs are inhibitory, these data suggest distinct roles for these neuronal classes in pain modulation. Along with direct excitation, we also found that presynaptic nAChRs enhanced GABAergic release preferentially onto neurons that lacked α7 nAChRs. In addition, presynaptic nAChRs enhanced glutamatergic inputs onto all PAG-RVM projection neuron classes to a similar extent. In behavioral testing, both systemic and intra-vlPAG administration of the α7 nAChR-selective agonist, PHA-543,613, was antinociceptive in the formalin assay. Furthermore, intra-vlPAG α7 antagonist pretreatment blocked PHA-543,613-induced antinociception via either administration method. Systemic administration of submaximal doses of the α7 agonist and morphine produced additive antinociceptive effects. Together, our findings indicate that the vlPAG is a key site of action for α7 nAChR-mediated antinociception.


Medulla Oblongata/drug effects , Neural Pathways/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Periaqueductal Gray/drug effects , Acetylcholine/pharmacology , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cholinergic Agents/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Gene Expression Regulation/drug effects , Male , Pain Measurement , Quinuclidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/metabolism , Receptors, Opioid, mu/metabolism , Synaptic Transmission/drug effects
5.
Neuropharmacology ; 117: 328-337, 2017 05 01.
Article En | MEDLINE | ID: mdl-28223211

Exposure to psychostimulants like cocaine or amphetamine leads to long-lasting sensitization of their behavioral and neurochemical effects. Here we characterized changes in AMPA receptor distribution and phosphorylation state in the rat nucleus accumbens (NAcc) weeks after amphetamine exposure to assess their potential contribution to sensitization by this drug. Using protein cross-linking, biochemical, subcellular fractionation, and slice electrophysiological approaches in the NAcc, we found that, unlike cocaine, previous exposure to amphetamine did not increase cell surface levels of either GluA1 or GluA2 AMPA receptor subunits, redistribution of these subunits to the synaptic or perisynaptic cellular membrane domains, protein-protein associations required to support the accumulation and retention of AMPA receptors in the PSD, or the peak amplitude of AMPA receptor mediated mEPSCs recorded in NAcc slices. On the other hand, exposure to amphetamine significantly slowed mEPSC decay times and increased levels in the PSD of PKA and CaMKII as well as phosphorylation by these kinases of the GluA1 S845 and S831 residues selectively in this cellular compartment. As the latter effects are known to respectively regulate channel open probability and duration as well as conductance, they provide a novel mechanism that could contribute to the long-lasting AMPA receptor dependent expression of sensitization by amphetamine. Rather than increase the number of surface and synaptic AMPA receptors as with cocaine, this mechanism could increase NAcc medium spiny neuron reactivity to glutamate afferents by increasing the phosphorylation state of critical regulatory sites in the AMPA receptor GluA1 subunit in the PSD.


Amphetamine/pharmacology , Cell Membrane/drug effects , Central Nervous System Stimulants/pharmacology , Nucleus Accumbens/drug effects , Receptors, AMPA/metabolism , Animals , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Male , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Nucleus Accumbens/metabolism , Phosphorylation/drug effects , Post-Synaptic Density/drug effects , Post-Synaptic Density/metabolism , Rats, Sprague-Dawley , Tissue Culture Techniques
6.
eNeuro ; 3(4)2016.
Article En | MEDLINE | ID: mdl-27517088

Ethanol (EtOH) and nicotine are the most widely coabused drugs. Tolerance to EtOH intoxication, including motor impairment, results in greater EtOH consumption and may result in a greater likelihood of addiction. Previous studies suggest that cross-tolerance between EtOH and nicotine may contribute to the abuse potential of these drugs. Here we demonstrate that repeated intermittent administration of either EtOH or nicotine in adult male Sprague Dawley rats results in tolerance to EtOH-induced motor impairment and increased EtOH self-administration. These findings suggest that nicotine and EtOH cross-tolerance results in decreased aversive and enhanced rewarding effects of EtOH. Endocannabinoid signaling in the dorsolateral striatum (DLS) has been implicated in both EtOH tolerance and reward, so we investigated whether nicotine or EtOH pretreatment might modulate endocannabinoid signaling in this region. Using similar EtOH and nicotine pretreatment methods resulted in increased paired-pulse ratios of evoked EPSCs in enkephalin-positive medium spiny neurons in DLS slices. Thus, EtOH and nicotine pretreatment may modulate glutamatergic synapses in the DLS presynaptically. Bath application of the CB1 receptor agonist Win 55,2-212 increased the paired-pulse ratio of evoked EPSCs in control slices, while Win 55,2-212 had no effect on paired-pulse ratio in slices from either EtOH- or nicotine-pretreated rats. Consistent with these effects, nicotine pretreatment occluded LTD induction by high-frequency stimulation of the corticostriatal inputs to the dorsolateral striatum. These results suggest that nicotine and EtOH pretreatment modulates striatal synapses to induce tolerance to the motor-impairing effects of EtOH, which may contribute to nicotine and EtOH coabuse.


Alcohol Drinking/physiopathology , Central Nervous System Depressants/administration & dosage , Corpus Striatum/drug effects , Ethanol/administration & dosage , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Alcohol Drinking/pathology , Animals , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Drug Tolerance , Enkephalins/metabolism , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/metabolism , Male , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Rats, Sprague-Dawley , Self Administration , Time Factors , Tissue Culture Techniques
7.
J Neurosci ; 36(29): 7768-78, 2016 07 20.
Article En | MEDLINE | ID: mdl-27445152

UNLABELLED: Nicotine and ethanol (EtOH) are among the most widely co-abused substances, and nicotinic acetylcholine receptors (nAChRs) contribute to the behavioral effects of both drugs. Along with their role in addiction, nAChRs also contribute to motor control circuitry. The α7 nAChR subtype is highly expressed in the laterodorsal tegmental nucleus (LDTg), a brainstem cholinergic center that contributes to motor performance through its projections to thalamic motor relay centers, including the mediodorsal thalamus. We demonstrate that EtOH concentrations just above the legal limits for intoxication in humans can inhibit α7 nAChRs in LDTg neurons from rats. This EtOH-induced inhibition is mediated by a decrease in cAMP/PKA signaling. The α7 nAChR-positive allosteric modulator PNU120596 [N-(5-chloro-2,4-dimethoxyphenyl)-N'-(5-methyl-3-isoxazolyl)-urea], which interferes with receptor desensitization, completely eliminated EtOH modulation of these receptors. These data suggest that EtOH inhibits α7 responses through a PKA-dependent enhancement of receptor desensitization. EtOH also inhibited the effects of nicotine at presynaptic α7 nAChRs on glutamate terminals in the mediodorsal thalamus. In vivo administration of PNU120596 either into the cerebral ventricles or directly into the mediodorsal thalamus attenuated EtOH-induced motor impairment. Thus, α7 nAChRs are likely important mediators of the motor impairing effects of moderate EtOH consumption. SIGNIFICANCE STATEMENT: The motor-impairing effects of ethanol contribute to intoxication-related injury and death. Here we explore the cellular and neural circuit mechanisms underlying ethanol-induced motor impairment. Physiologically relevant concentrations of ethanol inhibit activity of a nicotinic receptor subtype that is expressed in brain areas associated with motor control. That receptor inhibition is mediated by decreased receptor phosphorylation, suggesting an indirect modulation of cell signaling pathways to achieve the physiological effects.


Central Nervous System Depressants/toxicity , Ethanol/toxicity , Motor Disorders/chemically induced , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Cholinergic Agents/pharmacology , Cholinesterases/metabolism , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , In Vitro Techniques , Isoxazoles/pharmacology , Lysine/analogs & derivatives , Lysine/metabolism , Male , Neurotransmitter Agents/pharmacology , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Synaptic Potentials/drug effects
8.
J Neurosci ; 36(19): 5228-40, 2016 05 11.
Article En | MEDLINE | ID: mdl-27170121

UNLABELLED: Although dopamine receptor antagonism has long been associated with impairments in motor performance, more recent studies have shown that dopamine D2 receptor (D2R) antagonism, paired with a motor task, not only impairs motor performance concomitant with the pharmacodynamics of the drug, but also impairs future motor performance once antagonism has been relieved. We have termed this phenomenon "aberrant motor learning" and have suggested that it may contribute to motor symptoms in movement disorders such as Parkinson's disease (PD). Here, we show that chronic nicotine (cNIC), but not acute nicotine, treatment mitigates the acquisition of D2R-antagonist-induced aberrant motor learning in mice. Although cNIC mitigates D2R-mediated aberrant motor learning, cNIC has no effect on D1R-mediated motor learning. ß2-containing nicotinic receptors in dopamine neurons likely mediate the protective effect of cNIC against aberrant motor learning, because selective deletion of ß2 nicotinic subunits in dopamine neurons reduced D2R-mediated aberrant motor learning. Finally, both cNIC treatment and ß2 subunit deletion blunted postsynaptic responses to D2R antagonism. These results suggest that a chronic decrease in function or a downregulation of ß2-containing nicotinic receptors protects the striatal network against aberrant plasticity and aberrant motor learning induced by motor experience under dopamine deficiency. SIGNIFICANCE STATEMENT: Increasingly, aberrant plasticity and aberrant learning are recognized as contributing to the development and progression of movement disorders. Here, we show that chronic nicotine (cNIC) treatment or specific deletion of ß2 nicotinic receptor subunits in dopamine neurons mitigates aberrant motor learning induced by dopamine D2 receptor (D2R) blockade in mice. Moreover, both manipulations also reduced striatal dopamine release and blunt postsynaptic responses to D2R antagonists. These results suggest that chronic downregulation of function and/or receptor expression of ß2-containing nicotinic receptors alters presynaptic and postsynaptic striatal signaling to protect against aberrant motor learning. Moreover, these results suggest that cNIC treatment may alleviate motor symptoms and/or delay the deterioration of motor function in movement disorders by blocking aberrant motor learning.


Dopamine/deficiency , Learning/drug effects , Motor Activity , Nicotine/pharmacology , Receptors, Dopamine D2/metabolism , Receptors, Nicotinic/metabolism , Animals , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/physiology , Female , Male , Mice , Mice, Inbred C57BL , Nicotine/administration & dosage , Nicotinic Agonists/pharmacology , Synaptic Potentials
9.
Neurobiol Dis ; 76: 67-76, 2015 Apr.
Article En | MEDLINE | ID: mdl-25661301

Levodopa is the most effective therapy for the motor deficits of Parkinson's disease (PD), but long term treatment leads to the development of L-DOPA-induced dyskinesia (LID). Our previous studies indicate enhanced excitability of striatal cholinergic interneurons (ChIs) in mice expressing LID and reduction of LID when ChIs are selectively ablated. Recent gene expression analysis indicates that stimulatory H2 histamine receptors are preferentially expressed on ChIs at high levels in the striatum, and we tested whether a change in H2 receptor function might contribute to the elevated excitability in LID. Using two different mouse models of PD (6-hydroxydopamine lesion and Pitx3(ak/ak) mutation), we chronically treated the animals with either vehicle or l-DOPA to induce dyskinesia. Electrophysiological recordings indicate that histamine H2 receptor-mediated excitation of striatal ChIs is enhanced in mice expressing LID. Additionally, H2 receptor blockade by systemic administration of famotidine decreases behavioral LID expression in dyskinetic animals. These findings suggest that ChIs undergo a pathological change in LID with respect to histaminergic neurotransmission. The hypercholinergic striatum associated with LID may be dampened by inhibition of H2 histaminergic neurotransmission. This study also provides a proof of principle of utilizing selective gene expression data for cell-type-specific modulation of neuronal activity.


Cholinergic Neurons/physiology , Corpus Striatum/physiology , Dyskinesia, Drug-Induced/physiopathology , Parkinson Disease/complications , Receptors, Histamine H2/metabolism , Action Potentials , Animals , Cholinergic Neurons/metabolism , Corpus Striatum/metabolism , Dicyclomine/administration & dosage , Disease Models, Animal , Dyskinesia, Drug-Induced/metabolism , Famotidine/administration & dosage , Histamine H2 Antagonists/pharmacology , Interneurons/metabolism , Interneurons/physiology , Levodopa , Mice , Mice, Inbred C57BL
10.
Article En | MEDLINE | ID: mdl-25374536

The striatum plays a central role in motor control and motor learning. Appropriate responses to environmental stimuli, including pursuit of reward or avoidance of aversive experience all require functional striatal circuits. These pathways integrate synaptic inputs from limbic and cortical regions including sensory, motor and motivational information to ultimately connect intention to action. Although many neurotransmitters participate in striatal circuitry, one critically important player is acetylcholine (ACh). Relative to other brain areas, the striatum contains exceptionally high levels of ACh, the enzymes that catalyze its synthesis and breakdown, as well as both nicotinic and muscarinic receptor types that mediate its postsynaptic effects. The principal source of striatal ACh is the cholinergic interneuron (ChI), which comprises only about 1-2% of all striatal cells yet sends dense arbors of projections throughout the striatum. This review summarizes recent advances in our understanding of the factors affecting the excitability of these neurons through acute effects and long term changes in their synaptic inputs. In addition, we discuss the physiological effects of ACh in the striatum, and how changes in ACh levels may contribute to disease states during striatal dysfunction.

11.
J Neurosci ; 34(19): 6692-9, 2014 May 07.
Article En | MEDLINE | ID: mdl-24806695

Recent experimental evidence suggests that the low dopamine conditions in Parkinson's disease (PD) cause motor impairment through aberrant motor learning. Those data, along with computational models, suggest that this aberrant learning results from maladaptive corticostriatal plasticity and learned motor inhibition. Dopaminergic modulation of both corticostriatal long-term depression (LTD) and long-term potentiation (LTP) is proposed to be critical for these processes; however, the regulatory mechanisms underlying bidirectional corticostriatal plasticity are not fully understood. Previously, we demonstrated a key role for cAMP signaling in corticostriatal LTD. In this study, mouse brain slices were used to perform a parametric experiment that tested the impact of varying both intracellular cAMP levels and the strength of excitatory inputs on corticostriatal plasticity. Using slice electrophysiology in the dorsolateral striatum, we demonstrate that both LTP and LTD can be sequentially induced in the same D2-expressing neuron and that LTP was strongest with high intracellular cAMP and LFS, whereas LTD required low intracellular cAMP and high-frequency stimulation. Our results provide a molecular and cellular basis for regulating bidirectional corticostriatal synaptic plasticity and may help to identify novel therapeutic targets for blocking or reversing the aberrant synaptic plasticity that likely contributes to motor deficits in PD.


Cyclic AMP/physiology , Globus Pallidus/physiology , Neostriatum/physiology , Neuronal Plasticity/physiology , Neurons, Afferent/physiology , Synapses/physiology , Animals , Dopamine/physiology , Electric Stimulation , Electrophysiological Phenomena , Excitatory Postsynaptic Potentials/physiology , Female , Globus Pallidus/cytology , In Vitro Techniques , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neostriatum/cytology , Patch-Clamp Techniques
12.
J Neurophysiol ; 111(1): 103-11, 2014 Jan.
Article En | MEDLINE | ID: mdl-24089398

Nicotinic acetylcholine receptors (nAChRs) are expressed presynaptically on dopamine axon terminals, and their activation by endogenous acetylcholine from striatal cholinergic interneurons enhances dopamine release both independently of and in concert with dopamine neuron activity. Acute nAChR inactivation is believed to enhance the contrast between low- and high-frequency dopamine cell activity. Although these studies reveal a key role for acute activation and inactivation of nAChRs in striatal microcircuitry, it remains unknown if chronic inactivation/desensitization of nAChRs can alter dopamine release dynamics. Using in vivo cyclic voltammetry in anaesthetized mice, we examined whether chronic inactivation of nAChRs modulates dopamine release across a parametric range of stimulation, varying both frequency and pulse number. Deletion of ß2*nAChRs and chronic nicotine exposure greatly diminished dopamine release across the entire range of stimulation parameters. In addition, we observed a facilitation of dopamine release at low frequency and pulse number in wild-type mice that is absent in the ß2* knockout and chronic nicotine mice. These data suggest that deletion or chronic desensitization of nAChRs reduces the dynamic range of dopamine release in response to dopamine cell activity, decreasing rather than increasing contrast between high and low dopamine activity.


Dopamine/metabolism , Receptors, Nicotinic/metabolism , Substantia Nigra/metabolism , Action Potentials , Animals , Dopamine/pharmacology , Exocytosis , Mice , Mice, Inbred C57BL , Receptors, Nicotinic/genetics , Substantia Nigra/drug effects , Substantia Nigra/physiology
13.
Biochem Pharmacol ; 86(8): 1208-14, 2013 Oct 15.
Article En | MEDLINE | ID: mdl-23948066

Along with their well known role in nicotine addiction and autonomic physiology, neuronal nicotinic receptors (nAChRs) also have profound analgesic effects in animal models and humans. This is not a new idea, even in the early 1500s, soon after tobacco was introduced to the new world, its proponents listed pain relief among the beneficial properties of smoking. In recent years, analgesics that target specific nAChR subtypes have shown highly efficacious antinociceptive properties in acute and chronic pain models. To date, the side effects of these drugs have precluded their advancement to the clinic. This review summarizes the recent efforts to identify novel analgesics that target nAChRs, and outlines some of the key neural substrates that contribute to these physiological effects. There remain many unanswered mechanistic questions in this field, and there are still compelling reasons to explore neuronal nAChRs as targets for the relief of pain.


Analgesics/pharmacology , Cholinergic Agents/pharmacology , Receptors, Nicotinic/metabolism , Animals , Gene Expression Regulation/physiology , Humans , Pain/drug therapy , Pain/metabolism , Receptors, Nicotinic/genetics
14.
Eur J Neurosci ; 37(6): 1004-11, 2013 Mar.
Article En | MEDLINE | ID: mdl-23331514

Dopaminergic projections from the ventral tegmental area (VTA) to the nucleus accumbens (NAcc) mediate the behavioral and motivational effects of many drugs of abuse, including nicotine. Repeated intermittent administration of these drugs, a pattern often associated with initial drug exposure, sensitises the reactivity of dopamine (DA) neurons in this pathway, enhances the locomotor behaviors the drugs emit, and promotes their pursuit and self-administration. Here we show that activation of nicotinic acetylcholine receptors (nAChRs) in the VTA, but not the NAcc, is essential for the induction of locomotor sensitisation by nicotine. Repeated intermittent nicotine exposure (4 × 0.4 mg/kg, base, i.p., administered over 7 days), a regimen leading to long-lasting locomotor sensitisation, also produced upregulation of nAChRs in the VTA, but not the NAcc, in the hours following the last exposure injection. Functional nAChR upregulation was observed selectively in DA but not GABA neurons in the VTA. These effects were followed by long-term potentiation of excitatory inputs to these cells and increased nicotine-evoked DA overflow in the NAcc. Withdrawal symptoms were not observed following this exposure regimen. Thus, intermittent activation and upregulation by nicotine of nAChRs in DA neurons in the VTA may contribute to the development of behavioral sensitisation and increased liability for nicotine addiction.


Dopaminergic Neurons/physiology , Locomotion/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Ventral Tegmental Area/physiology , Animals , Central Nervous System Sensitization , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , GABAergic Neurons/metabolism , Long-Term Potentiation/drug effects , Male , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Up-Regulation , Ventral Tegmental Area/cytology , Ventral Tegmental Area/metabolism
15.
Cell Rep ; 2(6): 1747-61, 2012 Dec 27.
Article En | MEDLINE | ID: mdl-23246005

Dopamine contributes to corticostriatal plasticity and motor learning. Dopamine denervation profoundly alters motor performance, as in Parkinson's disease (PD); however, the extent to which these symptoms reflect impaired motor learning is unknown. Here, we demonstrate a D2 receptor blockade-induced aberrant learning that impedes future motor performance when dopamine signaling is restored, an effect diminished by coadministration of adenosine antagonists during blockade. We hypothesize that an inappropriate corticostriatal potentiation in striatopallidal cells of the indirect pathway underlies aberrant learning. We demonstrate synaptic potentiation in striatopallidal neurons induced by D2 blockade and diminished by application of an adenosine antagonist, consistent with behavioral observations. A neurocomputational model of the basal ganglia recapitulates the behavioral pattern and further links aberrant learning to plasticity in the indirect pathway. Thus, D2-mediated aberrant learning may contribute to motor deficits in PD, suggesting new avenues for the development of therapeutics.


Behavior, Animal , Dopamine/metabolism , Learning , Parkinson Disease , Signal Transduction , Adenosine/antagonists & inhibitors , Adenosine/metabolism , Animals , Dopamine D2 Receptor Antagonists , Mice , Mice, Transgenic , Motor Activity , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Receptors, Dopamine D2/metabolism
16.
J Neurophysiol ; 108(6): 1620-30, 2012 Sep.
Article En | MEDLINE | ID: mdl-22723669

Adolescence may be a period of vulnerability to drug addiction. In rats, elevated firing activity of ventral tegmental area (VTA) dopamine neurons predicts enhanced addiction liability. Our aim was to determine if dopamine neurons are more active in adolescents than in adults and to examine mechanisms underlying any age-related difference. VTA dopamine neurons fired faster in adolescents than in adults as measured with in vivo extracellular recordings. Dopamine neuron firing can be divided into nonbursting (single spikes) and bursting activity (clusters of high-frequency spikes). Nonbursting activity was higher in adolescents compared with adults. Frequency of burst events did not differ between ages, but bursts were longer in adolescents than in adults. Elevated dopamine neuron firing in adolescent rats was also observed in cell-attached recordings in ex vivo brain slices. Using whole cell recordings, we found that passive and active membrane properties were similar across ages. Hyperpolarization-activated cation currents and small-conductance calcium-activated potassium channel currents were also comparable across ages. We found no difference in dopamine D2-class autoreceptor function across ages, although the high baseline firing in adolescents resulted in autoreceptor activation being less effective at silencing neurons. Finally, AMPA receptor-mediated spontaneous excitatory postsynaptic currents occurred at lower frequency in adolescents; GABA(A) receptor-mediated spontaneous inhibitory postsynaptic currents occurred at both lower frequency and smaller amplitude in adolescents. In conclusion, VTA dopamine neurons fire faster in adolescence, potentially because GABA tone increases as rats reach adulthood. This elevation of firing rate during adolescence is consistent with it representing a vulnerable period for developing drug addiction.


Action Potentials , Dopaminergic Neurons/physiology , Ventral Tegmental Area/physiology , Age Factors , Animals , Excitatory Postsynaptic Potentials , Inhibitory Postsynaptic Potentials , Male , Rats , Rats, Sprague-Dawley , Receptors, AMPA/physiology , Receptors, Dopamine D2/physiology , Receptors, GABA/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Ventral Tegmental Area/growth & development
17.
J Neurol Sci ; 317(1-2): 66-73, 2012 Jun 15.
Article En | MEDLINE | ID: mdl-22459357

The cellular distribution of TAR DNA binding protein (TDP-43) is disrupted in several neurodegenerative disorders, including frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-U subtype) and amyotrophic lateral sclerosis (ALS). In these conditions, TDP-43 is found in neuronal cytoplasmic inclusions, with loss of the normal nuclear expression. The mechanisms leading to TDP-43 redistribution and its role in disease pathophysiology remain unknown. We describe an in vitro neural tissue model that reproduces TDP-43 relocalization and inclusion formation. Two week-old coronal organotypic mouse brain slice cultures were treated with tunicamycin for 7 days. In cortical regions of treated slice cultures, cytoplasmic inclusions of TDP-43 immunoreactivity were observed, with loss of nuclear TDP-43 immunoreactivity. These inclusions were found in both astrocytes and neurons, and were of both skein-like and round morphologies. In contrast, TDP-43 cytoplasmic inclusions were not found in slices treated with staurosporine to induce apoptosis, or with trans-4-carboxy-l-proline (PDC) to induce chronic glutamate excitotoxicity. Furthermore, TDP-43 cytoplasmic inclusions did not co-localize with cleaved caspase-3, suggesting that TDP-43 mislocalization does not generally accompany caspase activation or apoptosis. The induction of TDP-43 cytoplasmic translocation in cerebrocortical slice cultures by tunicamycin provides a platform for further mechanistic investigations of pathological processing of TDP-43.


Brain/metabolism , DNA-Binding Proteins/biosynthesis , Inclusion Bodies/metabolism , Inclusion Bodies/pathology , Tunicamycin/pharmacology , Animals , Animals, Newborn , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Brain/pathology , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Protein Transport/physiology , TDP-43 Proteinopathies/chemically induced , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology , Tunicamycin/toxicity
18.
J Neurophysiol ; 106(2): 801-8, 2011 Aug.
Article En | MEDLINE | ID: mdl-21593391

Tobacco use is a major public health problem, and although many smokers report that they want to quit, only a small percentage succeed. Side effects associated with nicotine withdrawal, including depression, anxiety, and restlessness, certainly contribute to the low success rate. The dorsal raphe nucleus (DRN) is a serotonergic center with many functions, including control of mood and emotional state. We investigated the effect of nicotine on DRN neurons that project to the nucleus accumbens (NAc), an area involved in reward-related behaviors. Using a retrograde labeling method, we found that 75% of DRN-NAc projection neurons are serotonergic. In coronal slices that include the DRN, whole cell recordings were conducted on neurons identified by fluorescent backlabeling from NAc or randomly selected within the nucleus. Nicotine increased action potential firing rates in a subset of DRN neurons. Voltage-clamp recording revealed nicotinic acetylcholine receptor (nAChR)-mediated inward currents that contribute to the nicotine-induced excitation. Nicotinic receptors also indirectly affect excitability by modulating synaptic inputs to these neurons. Nicotine enhanced excitatory glutamatergic inputs to a subset of DRN-NAc projection neurons, while inhibitory γ-aminobutyric acid (GABA)ergic inputs were modulated either positively or negatively in a subset of these neurons. The net effect of nAChR activation is enhancement of serotonergic output from DRN to the NAc, which may contribute to the effects of nicotine on mood and affect.


Excitatory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/physiology , Nicotine/pharmacology , Nucleus Accumbens/physiology , Raphe Nuclei/physiology , Serotonergic Neurons/physiology , Animals , Excitatory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/drug effects , Male , Nucleus Accumbens/drug effects , Organ Culture Techniques , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Serotonergic Neurons/drug effects
19.
J Neurosci ; 31(18): 6710-20, 2011 May 04.
Article En | MEDLINE | ID: mdl-21543600

Drug-induced changes in synaptic strength are hypothesized to contribute to appetitive behavior and addiction. Nicotine, the major addictive substance in tobacco, activates nicotinic receptors (nAChRs) to initiate a series of adaptive changes at the cellular and circuit levels in brain, particularly the ventral tegmental area (VTA). Our laboratory previously reported that nicotine facilitates induction of long-term potentiation (LTP) in VTA dopamine (DA) neurons by increasing glutamate release via activation of α7 nAChRs on the glutamate terminals, suggesting a critical presynaptic contribution of nicotine in LTP induction. In the present study, we used an in vitro exposure paradigm to study the effect of nicotine on excitatory synaptic strength. Brief exposure of nicotine to brain slices from drug-naive adult rats followed by a period of recovery resulted in an NMDA receptor (NMDAR)-dependent increase of AMPA receptor/NMDAR ratio in VTA DA neurons, which is consistent with the induction of LTP. These effects are similar to that induced by a single in vivo nicotine injection intraperitoneally. The induction of synaptic potentiation required excitation of DA neurons mediated by somatodendritic α4ß2 nAChRs, as well as enhancement of NMDAR function via D(5) dopamine receptors, also on DA neurons. Nicotine-induced increase of presynaptic glutamate release also contributed to the induction of synaptic plasticity, likely through increased activation of NMDAR. These results identified important receptor systems involved in nicotine-induced long-term changes in excitatory synaptic input to VTA DA neurons. The data also revealed remarkable similarity in the mechanisms underlying synaptic plasticity induced by nicotine and cocaine in the VTA.


Dopamine/metabolism , Long-Term Potentiation/drug effects , Neurons/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Ventral Tegmental Area/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Analysis of Variance , Animals , Electrophysiology , Immunohistochemistry , Long-Term Potentiation/physiology , Male , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Nicotinic/metabolism , Synapses/drug effects , Synapses/physiology , Ventral Tegmental Area/physiology
20.
Proc Natl Acad Sci U S A ; 108(2): 840-5, 2011 Jan 11.
Article En | MEDLINE | ID: mdl-21187382

Treatment of Parkinson disease (PD) with L-3,4-dihydroxyphenylalanine (L-DOPA) dramatically relieves associated motor deficits, but L-DOPA-induced dyskinesias (LID) limit the therapeutic benefit over time. Previous investigations have noted changes in striatal medium spiny neurons, including abnormal activation of extracellular signal-regulated kinase1/2 (ERK). Using two PD models, the traditional 6-hydroxydopamine toxic lesion and a genetic model with nigrostriatal dopaminergic deficits, we found that acute dopamine challenge induces ERK activation in medium spiny neurons in denervated striatum. After repeated L-DOPA treatment, however, ERK activation diminishes in medium spiny neurons and increases in striatal cholinergic interneurons. ERK activation leads to enhanced basal firing rate and stronger excitatory responses to dopamine in striatal cholinergic neurons. Pharmacological blockers of ERK activation inhibit L-DOPA-induced changes in ERK phosphorylation, neuronal excitability, and the behavioral manifestation of LID. In addition, a muscarinic receptor antagonist reduces LID. These data indicate that increased dopamine sensitivity of striatal cholinergic neurons contributes to the expression of LID, which suggests novel therapeutic targets for LID.


Cholinergic Fibers/metabolism , Dyskinesias/metabolism , Gene Expression Regulation , Levodopa/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Adenosine A2 Receptor Antagonists/chemistry , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , Aphakia/metabolism , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Dopamine/genetics , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Phosphorylation , Transcription Factors/genetics
...