Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Eur J Neurosci ; 49(6): 859-868, 2019 03.
Article in English | MEDLINE | ID: mdl-29923650

ABSTRACT

L-dopa induced dyskinesias (LIDs) are a disabling motor complication of L-dopa therapy for Parkinson's disease (PD) management. Treatment options remain limited and the underlying network mechanisms remain unclear due to a complex pathophysiology. What is well-known, however, is that aberrant striatal signaling plays a key role in LIDs development. Here, we discuss the specific contribution of striatal cholinergic interneurons (ChIs) and GABAergic medium spiny projection neurons (MSNs) with a particular focus on how cholinergic signaling may integrate multiple striatal systems to modulate LIDs expression. Enhanced ChI transmission, altered MSN activity and the associated abnormal downstream signaling responses that arise with nigrostriatal damage are well known to contribute to LIDs development. In fact, enhancing M4 muscarinic receptor activity, a receptor favorably expressed on D1 dopamine receptor-expressing MSNs dampens their activity to attenuate LIDs. Likewise, ChI activation via thalamostriatal neurons is shown to interrupt cortical signaling to enhance D2 dopamine receptor-expressing MSN activity via M1 muscarinic receptors, which may interrupt ongoing motor activity. Notably, numerous preclinical studies also show that reducing nicotinic cholinergic receptor activity decreases LIDs. Taken together, these studies indicate the importance of cholinergic control of striatal neuronal activity and point to muscarinic and nicotinic receptors as significant pharmacological targets for alleviating LIDs in PD patients.


Subject(s)
Cholinergic Agents/pharmacology , Corpus Striatum/drug effects , Levodopa/pharmacology , Parkinson Disease/drug therapy , Aged , Cholinergic Neurons/metabolism , Corpus Striatum/metabolism , Female , Humans , Hypokinesia/physiopathology , Male , Middle Aged , Neurons/metabolism , Neurons/physiology , Parkinson Disease/physiopathology
2.
Mov Disord ; 32(4): 538-548, 2017 04.
Article in English | MEDLINE | ID: mdl-28256010

ABSTRACT

BACKGROUND: Dyskinesias are a disabling motor complication that arises with prolonged l-dopa treatment. Studies using D1 receptor drugs and genetically modified mice suggest that medium spiny neurons expressing D1 receptors play a primary role in l-dopa-induced dyskinesias. However, the specific role of these neurons in dyskinesias is not fully understood. METHODS: We used optogenetics, which allows for precise modulation of select neurons in vivo, to investigate whether striatal D1-expressing medium spiny neuron activity regulates abnormal involuntary movements or dyskinesia in parkinsonian mice. D1-cre mice unilaterally lesioned with 6-hydroxydopamine received striatal injections of cre-dependent channelrhodopsin2 virus or control virus. After stable virus expression, the effect of optical stimulation on dyskinesia was tested in l-dopa-naïve and l-dopa-primed mice. RESULTS: Single-pulse and burst-optical stimulation of D1-expressing medium spiny neurons induced dyskinesias in l-dopa-naïve channelrhodopsin2 mice. In stably dyskinetic mice, l-dopa injection induced dyskinesia to a similar or somewhat greater extent than optical stimulation. Combined l-dopa administration and stimulation resulted in an additive increase in dyskinesias, indicating that other mechanisms also contribute. Molecular studies indicate that changes in extracellular signal-regulated kinase phosphorylation in D1-expressing medium spiny neurons are involved. Optical stimulation did not ameliorate parkinsonism in l-dopa-naïve mice. However, it improved parkinsonism in l-dopa-primed mice to a similar extent as l-dopa administration. None of the stimulation paradigms enhanced dyskinesia or modified parkinsonism in l-dopa-naïve or l-dopa-primed control virus mice. CONCLUSION: The data provide direct evidence that striatal D1-expressing medium spiny neuron stimulation is sufficient to induce dyskinesias and contributes to the regulation of motor control. © 2017 International Parkinson and Movement Disorder Society.


Subject(s)
Corpus Striatum/cytology , Corpus Striatum/metabolism , Dyskinesias/etiology , Neurons/physiology , Parkinsonian Disorders/pathology , Receptors, Dopamine D1/metabolism , Animals , Antiparkinson Agents/adverse effects , Channelrhodopsins , Cocaine/analogs & derivatives , Cocaine/pharmacokinetics , Corpus Striatum/diagnostic imaging , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/metabolism , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/radiation effects , Levodopa/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Receptors, Dopamine D1/genetics , Sympatholytics/toxicity
3.
Exp Neurol ; 286: 32-39, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27658674

ABSTRACT

Tardive dyskinesia (TD) is a drug-induced movement disorder that arises with antipsychotics. These drugs are the mainstay of treatment for schizophrenia and bipolar disorder, and are also prescribed for major depression, autism, attention deficit hyperactivity, obsessive compulsive and post-traumatic stress disorder. There is thus a need for therapies to reduce TD. The present studies and our previous work show that nicotine administration decreases haloperidol-induced vacuous chewing movements (VCMs) in rodent TD models, suggesting a role for the nicotinic cholinergic system. Extensive studies also show that D2 dopamine receptors are critical to TD. However, the precise involvement of striatal cholinergic interneurons and D2 medium spiny neurons (MSNs) in TD is uncertain. To elucidate their role, we used optogenetics with a focus on the striatum because of its close links to TD. Optical stimulation of striatal cholinergic interneurons using cholineacetyltransferase (ChAT)-Cre mice expressing channelrhodopsin2-eYFP decreased haloperidol-induced VCMs (~50%), with no effect in control-eYFP mice. Activation of striatal D2 MSNs using Adora2a-Cre mice expressing channelrhodopsin2-eYFP also diminished antipsychotic-induced VCMs, with no change in control-eYFP mice. In both ChAT-Cre and Adora2a-Cre mice, stimulation or mecamylamine alone similarly decreased VCMs with no further decline with combined treatment, suggesting nAChRs are involved. Striatal D2 MSN activation in haloperidol-treated Adora2a-Cre mice increased c-Fos+ D2 MSNs and decreased c-Fos+ non-D2 MSNs, suggesting a role for c-Fos. These studies provide the first evidence that optogenetic stimulation of striatal cholinergic interneurons and GABAergic MSNs modulates VCMs, and thus possibly TD. Moreover, they suggest nicotinic receptor drugs may reduce antipsychotic-induced TD.


Subject(s)
Cholinergic Neurons/physiology , Corpus Striatum/pathology , GABAergic Neurons/physiology , Tardive Dyskinesia/pathology , Animals , Antipsychotic Agents/toxicity , Channelrhodopsins , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Haloperidol/toxicity , Male , Mastication/drug effects , Mecamylamine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nicotine/therapeutic use , Nicotinic Agonists/therapeutic use , Nicotinic Antagonists/pharmacology , Receptors, Dopamine D2/metabolism , Tardive Dyskinesia/chemically induced , Tardive Dyskinesia/drug therapy
4.
Neurobiol Dis ; 91: 47-58, 2016 07.
Article in English | MEDLINE | ID: mdl-26921469

ABSTRACT

L-dopa-induced dyskinesias (LIDs) are a serious complication of L-dopa therapy for Parkinson's disease. Emerging evidence indicates that the nicotinic cholinergic system plays a role in LIDs, although the pathways and mechanisms are poorly understood. Here we used optogenetics to investigate the role of striatal cholinergic interneurons in LIDs. Mice expressing cre-recombinase under the control of the choline acetyltransferase promoter (ChAT-Cre) were lesioned by unilateral injection of 6-hydroxydopamine. AAV5-ChR2-eYFP or AAV5-control-eYFP was injected into the dorsolateral striatum, and optical fibers implanted. After stable virus expression, mice were treated with L-dopa. They were then subjected to various stimulation protocols for 2h and LIDs rated. Continuous stimulation with a short duration optical pulse (1-5ms) enhanced LIDs. This effect was blocked by the general muscarinic acetylcholine receptor (mAChR) antagonist atropine indicating it was mAChR-mediated. By contrast, continuous stimulation with a longer duration optical pulse (20ms to 1s) reduced LIDs to a similar extent as nicotine treatment (~50%). The general nicotinic acetylcholine receptor (nAChR) antagonist mecamylamine blocked the decline in LIDs with longer optical pulses showing it was nAChR-mediated. None of the stimulation regimens altered LIDs in control-eYFP mice. Lesion-induced motor impairment was not affected by optical stimulation indicating that cholinergic transmission selectively regulates LIDs. Longer pulse stimulation increased the number of c-Fos expressing ChAT neurons, suggesting that changes in this immediate early gene may be involved. These results demonstrate that striatal cholinergic interneurons play a critical role in LIDs and support the idea that nicotine treatment reduces LIDs via nAChR desensitization.


Subject(s)
Corpus Striatum/drug effects , Dyskinesia, Drug-Induced/metabolism , Interneurons/drug effects , Levodopa/pharmacology , Neostriatum/drug effects , Nicotine/pharmacology , Animals , Choline O-Acetyltransferase/metabolism , Corpus Striatum/metabolism , Interneurons/metabolism , Mice , Neostriatum/metabolism , Nicotinic Agonists/pharmacology , Optogenetics/methods
5.
Mov Disord ; 30(14): 1901-1911, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26573698

ABSTRACT

BACKGROUND: ABT-126 is a novel, safe, and well-tolerated α7 nicotinic receptor agonist in a Phase 2 Alzheimer's disease study. We tested the antidyskinetic effect of ABT-126 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated squirrel monkeys with moderate and more severe nigrostriatal damage. METHODS: Monkeys (n = 21, set 1) were lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine 1-2×. When parkinsonian, they were gavaged with levodopa (10 mg/kg)/carbidopa (2.5 mg/kg) twice daily and dyskinesias rated. They were then given nicotine in drinking water (n = 5), or treated with vehicle (n = 6) or ABT-126 (n = 10) twice daily orally 30 min before levodopa. Set 1 was then re-lesioned 1 to 2 times for a total of 3 to 4 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine injections. The antidyskinetic effect of ABT-126, nicotine, and the ß2* nicotinic receptor agonist ABT-894 was re-assessed. Another group of monkeys (n = 23, set 2) were lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine only 1× to 2×. They were treated with levodopa/carbidopa, administered the α7 agonist ABT-107 (n = 6), ABT-894 (n = 6), nicotine (n = 5), or vehicle (n = 6) and dyskinesias evaluated. All monkeys were euthanized and the dopamine transporter measured. RESULTS: With moderate nigrostriatal damage (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine 1×-2×), ABT-126 dose-dependently decreased dyskinesias (∼60%), with similar results seen with ABT-894 (∼60%) or nicotine (∼60%). With more severe damage (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine 3-4×), ABT-126 and nicotine reduced dyskinesias, but ABT-894 did not. The dopamine transporter was 41% and 8.9% of control, with moderate and severe nigrostriatal damage, respectively. No drug modified parkinsonism. CONCLUSION: The novel α7 nicotinic receptor drug ABT-126 reduced dyskinesias in monkeys with both moderate and severe nigrostriatal damage. ABT-126 may be useful to reduce dyskinesias in both early- and later-stage Parkinson's disease.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Levodopa/adverse effects , Nicotinic Agonists/therapeutic use , Parkinsonian Disorders/drug therapy , Substantia Nigra/pathology , alpha7 Nicotinic Acetylcholine Receptor/agonists , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antiparkinson Agents/therapeutic use , Dyskinesia, Drug-Induced/pathology , Female , Levodopa/therapeutic use , Male , Nicotinic Agonists/pharmacology , Parkinsonian Disorders/pathology , Saimiri
6.
Neuropsychol Rev ; 25(4): 371-83, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26553323

ABSTRACT

One of the primary deficits in Parkinson's disease (PD) is the loss of dopaminergic neurons in the substantia nigra pars compacta which leads to striatal dopaminergic deficits that underlie the motor symptoms associated with the disease. A plethora of animal models have been developed over the years to uncover the molecular alterations that lead to PD development. These models have provided valuable information on neurotransmitter pathways and mechanisms involved. One such a system is the nicotinic cholinergic system. Numerous studies show that nigrostriatal damage affects nicotinic receptor-mediated dopaminergic signaling; therefore therapeutic modulation of the nicotinic cholinergic system may offer a novel approach to manage PD. In fact, there is evidence showing that nicotinic receptor drugs may be useful as neuroprotective agents to prevent Parkinson's disease progression. Additional preclinical studies also show that nicotinic receptor drugs may be beneficial for the treatment of L-dopa induced dyskinesias. Here, we review preclinical findings supporting the idea that nicotinic receptors are valuable therapeutic targets for PD.


Subject(s)
Antiparkinson Agents/pharmacology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/metabolism , Receptors, Nicotinic/metabolism , Animals , Antiparkinson Agents/therapeutic use , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
7.
Int Rev Neurobiol ; 124: 247-71, 2015.
Article in English | MEDLINE | ID: mdl-26472532

ABSTRACT

Parkinson's disease is a progressive neurodegenerative disorder associated with tremor, rigidity, and bradykinesia, as well as nonmotor symptoms including autonomic impairments, olfactory dysfunction, sleep disturbances, depression, and dementia. Although the major neurological deficit is a loss of nigrostriatal dopaminergic neurons, multiple neurotransmitters systems are compromised in Parkinson's disease. Consistent with this observation, dopamine replacement therapy dramatically improves Parkinson's disease motor symptoms. Additionally, drugs targeting the serotonergic, glutamatergic, adenosine, and other neurotransmitter systems may be beneficial. Recent evidence also indicates that nicotinic cholinergic drugs may be useful for the management of Parkinson's disease. This possibility initially arose from the results of epidemiological studies, which showed that smoking was associated with a decreased incidence of Parkinson's disease, an effect mediated in part by the nicotine in smoke. Further evidence for this idea stemmed from preclinical studies which showed that nicotine administration reduced nigrostriatal damage in parkinsonian rodents and monkeys. In addition to a potential neuroprotective role, emerging work indicates that nicotinic receptor drugs improve the abnormal involuntary movements or dyskinesias that arise as a side effect of l-dopa treatment, the gold standard therapy for Parkinson's disease. Both nicotine and nicotinic receptor drugs reduced l-dopa-induced dyskinesias by over 50% in parkinsonian rodent and monkey models. Notably, nicotine also attenuated the abnormal involuntary movements or tardive dyskinesias that arise with antipsychotic treatment. These observations, coupled with reports that nicotinic receptor drugs have procognitive and antidepressant effects, suggest that central nervous system (CNS) nicotinic receptors may represent useful targets for the treatment of movement disorders.


Subject(s)
Antiparkinson Agents/therapeutic use , Movement Disorders/etiology , Nicotine/therapeutic use , Parkinson Disease/drug therapy , Antiparkinson Agents/adverse effects , Humans
8.
Pharmacol Res Perspect ; 3(1): e00105, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25692023

ABSTRACT

An important factor contributing to the high relapse rates among smokers is nicotine withdrawal symptoms. Multiple studies suggest that decreased dopamine release in nucleus accumbens plays a key role in withdrawal. However, recent reports showed that long-term nicotine exposure itself also decreases accumbal dopamine release, suggesting that additional mechanisms are involved in withdrawal. Here, we used real-time cyclic voltammetry in brain slices containing the nucleus accumbens to further elucidate the changes in dopamine release linked to nicotine withdrawal. Rats received vehicle or nicotine via the drinking water for 2-3 months. Studies assessing the expression of somatic signs in vehicle-treated, nicotine-treated, and 24-h nicotine withdrawn rats showed that nicotine withdrawal led to a significant increase in somatic signs. Subsequent voltammetry studies showed that long-term nicotine decreased single-pulse-stimulated dopamine release via an interaction at α6ß2* receptors. Nicotine withdrawal led to a partial recovery in α6ß2* receptor-mediated release. In addition, long-term nicotine treatment alone increased dopamine release paired-pulse ratios and this was partially reversed with nicotine removal. We then evaluated the effect of bath-applied nicotine and varenicline on dopamine release. Nicotine and varenicline both decreased single-pulse-stimulated release in vehicle-treated, nicotine-treated, and nicotine withdrawn rats. However, bath-applied varenicline increased paired-pulse ratios to a greater extent than nicotine during long-term nicotine treatment and after its withdrawal. Altogether these data suggest that nicotine withdrawal is associated with a partial restoration of dopamine release measures to control levels and that varenicline's differential modulation of dopamine release may contribute to its mechanism of action.

9.
Pharmacol Ther ; 144(1): 50-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24836728

ABSTRACT

A large body of evidence using experimental animal models shows that the nicotinic cholinergic system is involved in the control of movement under physiological conditions. This work raised the question whether dysregulation of this system may contribute to motor dysfunction and whether drugs targeting nicotinic acetylcholine receptors (nAChRs) may be of therapeutic benefit in movement disorders. Accumulating preclinical studies now show that drugs acting at nAChRs improve drug-induced dyskinesias. The general nAChR agonist nicotine, as well as several nAChR agonists (varenicline, ABT-089 and ABT-894), reduces l-dopa-induced abnormal involuntary movements or dyskinesias up to 60% in parkinsonian nonhuman primates and rodents. These dyskinesias are potentially debilitating abnormal involuntary movements that arise as a complication of l-dopa therapy for Parkinson's disease. In addition, nicotine and varenicline decrease antipsychotic-induced abnormal involuntary movements in rodent models of tardive dyskinesia. Antipsychotic-induced dyskinesias frequently arise as a side effect of chronic drug treatment for schizophrenia, psychosis and other psychiatric disorders. Preclinical and clinical studies also show that the nAChR agonist varenicline improves balance and coordination in various ataxias. Lastly, nicotine has been reported to attenuate the dyskinetic symptoms of Tourette's disorder. Several nAChR subtypes appear to be involved in these beneficial effects of nicotine and nAChR drugs including α4ß2*, α6ß2* and α7 nAChRs (the asterisk indicates the possible presence of other subunits in the receptor). Overall, the above findings, coupled with nicotine's neuroprotective effects, suggest that nAChR drugs have potential for future drug development for movement disorders.


Subject(s)
Movement Disorders/drug therapy , Nicotine/pharmacology , Receptors, Nicotinic/drug effects , Animals , Antiparkinson Agents/adverse effects , Antipsychotic Agents/adverse effects , Drug Design , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/physiopathology , Humans , Movement Disorders/physiopathology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism
10.
J Neurochem ; 127(6): 762-71, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23992036

ABSTRACT

Long-term nicotine exposure induces alterations in dopamine transmission in nucleus accumbens that sustain the reinforcing effects of smoking. One approach to understand the adaptive changes that arise involves measurement of endogenous dopamine release using voltammetry. We therefore treated rats for 2-3 months with nicotine and examined alterations in nAChR subtype expression and electrically evoked dopamine release in rat nucleus accumbens shell, a region key in addiction. Long-term nicotine treatment selectively decreased stimulated α6ß2* nAChR-mediated dopamine release compared with vehicle-treated rats. It also reduced α6ß2* nAChRs, suggesting the receptor decline may contribute to the functional loss. This decreased response in release after chronic nicotine treatment was still partially sensitive to the agonist nicotine. Studies with an acetylcholinesterase inhibitor demonstrated that the response was also sensitive to increased endogenous acetylcholine. However, unlike the agonists, nAChR antagonists decreased dopamine release only in vehicle- but not nicotine-treated rats. As antagonists function by blocking the action of acetylcholine, their ineffectiveness suggests that reduced acetylcholine levels partly underlie the dampened α6ß2* nAChR-mediated function in nicotine-treated rats. As long-term nicotine modifies dopamine release by decreasing α6ß2* nAChRs and their function, these data suggest that interventions that target this subtype may be useful for treating nicotine dependence. Long-term nicotine treatment decreases dopamine (DA) transmission in the mesolimbic dopaminergic system. Our data suggest this may involve a decrease in α6ß2* nicotinic receptor expression and function. These changes may play a key role in nicotine reward and dependence.


Subject(s)
Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nucleus Accumbens/drug effects , Receptors, Nicotinic/metabolism , Ambenonium Chloride/pharmacology , Animals , Cholinesterase Inhibitors/pharmacology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Down-Regulation , Electric Stimulation , Male , Nicotinic Antagonists/pharmacology , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
11.
Neurobiol Dis ; 50: 30-41, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23009753

ABSTRACT

L-dopa-induced dyskinesias (LIDs) are abnormal involuntary movements that develop with long term L-dopa therapy for Parkinson's disease. Studies show that nicotine administration reduced LIDs in several parkinsonian animal models. The present work was done to understand the factors that regulate the nicotine-mediated reduction in LIDs in MPTP-lesioned nonhuman primates. To approach this, we used two groups of monkeys, one with mild-moderate and the other with more severe parkinsonism rendered dyskinetic using L-dopa. In mild-moderately parkinsonian monkeys, nicotine pretreatment (300 µg/ml via drinking water) prevented the development of LIDs by ~75%. This improvement was maintained when the nicotine dose was lowered to 50 µg/ml but was lost with nicotine removal. Nicotine re-exposure again decreased LIDs. By contrast, nicotine treatment did not reduce LIDs in monkeys with more severe parkinsonism. We next determined how nicotine's ability to reduce LIDs correlated with lesion-induced changes in the striatal dopamine transporter and (3)H-dopamine release in these two groups of monkeys. The striatal dopamine transporter was reduced to 54% and 28% of control in mild-moderately and more severely parkinsonian monkeys, respectively. However, basal, K(+), α4ß2* and α6ß2* nAChR-evoked (3)H-dopamine release were near control levels in striatum of mild-moderately parkinsonian monkeys. By contrast, these same release measures were reduced to a significantly greater extent in striatum of more severely parkinsonian monkeys. Thus, nicotine best improves LIDs in lesioned monkeys in which striatal dopamine transmission is still relatively intact. These data suggest that nicotine treatment would most effectively reduce LIDs in patients with mild to moderate Parkinson's disease.


Subject(s)
Dopamine/metabolism , Dyskinesia, Drug-Induced/metabolism , Dyskinesia, Drug-Induced/prevention & control , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine Agonists/adverse effects , Female , Levodopa/adverse effects , MPTP Poisoning/drug therapy , Male , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Saimiri
12.
Article in English | MEDLINE | ID: mdl-22679419

ABSTRACT

We have previously demonstrated that Type I neuronal nitric oxide synthase (nNOS)-expressing neurons are sleep-active in the cortex of mice, rats, and hamsters. These neurons are known to be GABAergic, to express Neuropeptide Y (NPY) and, in rats, to co-express the Substance P (SP) receptor NK1, suggesting a possible role for SP in sleep/wake regulation. To evaluate the degree of co-expression of nNOS and NK1 in the cortex among mammals, we used double immunofluorescence for nNOS and NK1 and determined the anatomical distribution in mouse, rat, and squirrel monkey cortex. Type I nNOS neurons co-expressed NK1 in all three species although the anatomical distribution within the cortex was species-specific. We then performed in vitro patch clamp recordings in cortical neurons in mouse and rat slices using the SP conjugate tetramethylrhodamine-SP (TMR-SP) to identify NK1-expressing cells and evaluated the effects of SP on these neurons. Bath application of SP (0.03-1 µM) resulted in a sustained increase in firing rate of these neurons; depolarization persisted in the presence of tetrodotoxin. These results suggest a conserved role for SP in the regulation of cortical sleep-active neurons in mammals.

13.
Mov Disord ; 27(8): 947-57, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22693036

ABSTRACT

Converging research efforts suggest that nicotine and other drugs that act at nicotinic acetylcholine receptors (nAChRs) may be beneficial in the management of Parkinson's disease. This idea initially stemmed from the results of epidemiological studies that demonstrated that smoking is associated with a decreased incidence of Parkinson's disease. The subsequent finding that nicotine administration protected against nigrostriatal damage in parkinsonian animal models led to the idea that nicotine in tobacco products may contribute to this apparent protective action. Nicotine most likely exerts its effects by interacting at nAChRs. Accumulating research indicates that multiple subtypes containing nAChRs, including α4ß2, α6ß2, and/or α7, may be involved. Stimulation of nAChRs initially activates various intracellular transduction pathways primarily via alterations in calcium signaling. Consequent adaptations in immune responsiveness and trophic factors may ultimately mediate nicotine's ability to reduce/halt the neuronal damage that arises in Parkinson's disease. In addition to a potential neuroprotective action, nicotine also has antidepressant properties and improves attention/cognition. Altogether, these findings suggest that nicotine and nAChR drugs represent promising therapeutic agents for the management of Parkinson's disease.


Subject(s)
Neuroprotective Agents , Nicotine/therapeutic use , Nicotinic Agonists/therapeutic use , Parkinson Disease/prevention & control , Animals , Corpus Striatum/pathology , Disease Models, Animal , Humans , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/prevention & control , Receptors, Nicotinic/drug effects , Signal Transduction/drug effects , Smoking/epidemiology , Substantia Nigra/pathology
14.
J Pharmacol Exp Ther ; 342(2): 335-44, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22562772

ABSTRACT

Tobacco use is a leading cause of preventable deaths worldwide. However, current smoking cessation therapies have very limited long-term success rates. Considerable research effort is therefore focused on identification of central nervous system changes with nicotine exposure because this may lead to more successful treatment options. Although recent work suggests that α6ß2* nicotinic acetylcholine receptors (nAChRs) play a dominant role in dopaminergic function in rodent nucleus accumbens, the effects of long-term nicotine exposure remain to be determined. Here, we used cyclic voltammetry to investigate α6ß2* nAChR-mediated release with long-term nicotine treatment in nonhuman primate nucleus accumbens shell. Control studies showed that nAChR-mediated dopamine release occurs predominantly through the α6ß2* receptor subtype. Unexpectedly, there was a complete loss of α6ß2* nAChR-mediated activity after several months of nicotine treatment. This decline in function was observed with both single- and multiple-pulse-stimulated dopamine release. Paired-pulse studies showed that the facilitation of dopamine release with multiple pulsing observed in controls in the presence of nAChR antagonist was lost with long-term nicotine treatment. Nicotine-evoked [(3)H]dopamine release from nucleus accumbens synaptosomes was similar in nicotine- and vehicle-treated monkeys, indicating that long-term nicotine administration does not directly modify α6ß2* nAChR-mediated dopamine release. Dopamine uptake rates, as well as dopamine transporter and α6ß2* nAChRs levels, were also not changed with nicotine administration. These data indicate that nicotine exposure, as occurs with smoking, has major effects on cellular mechanisms linked to α6ß2* nAChR-mediated dopamine release and that this receptor subtype may represent a novel therapeutic target for smoking cessation.


Subject(s)
Dopamine/administration & dosage , Dopamine/metabolism , Nicotine/pharmacology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Animals , Biological Transport/drug effects , Female , Male , Receptors, Nicotinic/metabolism , Saimiri , Smoking Cessation , Synaptosomes/drug effects , Synaptosomes/metabolism
15.
Biochem Pharmacol ; 82(8): 873-82, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21684266

ABSTRACT

Although a relative newcomer to the nicotinic acetylcholine receptor (nAChR) family, substantial evidence suggests that α6 containing nAChRs play a key role in CNS function. This subtype is unique in its relatively restricted localization to the visual system and catecholaminergic pathways. These latter include the mesolimbic and nigrostriatal dopaminergic systems, which may account for the involvement of α6 containing nAChRs in the rewarding properties of nicotine and in movement. Here, we review the literature on the role of α6 containing nAChRs with a focus on the striatum and nucleus accumbens. This includes molecular, electrophysiological and behavioral studies in control and lesioned animal models, as well as in different genetic models. Converging evidence suggest that the major α6 containing nAChRs subtypes in the nigrostriatal and mesolimbic dopamine system are the α6ß2ß3 and α6α4ß2ß3 nAChR populations. They appear to have a dominant role in regulating dopamine release, with consequent effects on nAChR-modulated dopaminergic functions such as reinforcement and motor behavior. Altogether these data suggest that drugs directed to α6 containing nAChRs may be of benefit for the treatment of addiction and for neurological disorders with locomotor deficits such as Parkinson's disease.


Subject(s)
Central Nervous System/metabolism , Dopamine/metabolism , Parkinson Disease/metabolism , Receptors, Nicotinic/physiology , Substance-Related Disorders/metabolism , Animals , Corpus Striatum/metabolism , Humans , Nucleus Accumbens/metabolism , Protein Subunits , Receptors, Nicotinic/metabolism , Visual Cortex/metabolism
16.
Mol Cell Pharmacol ; 3(1): 1-6, 2011.
Article in English | MEDLINE | ID: mdl-21499569

ABSTRACT

L-dopa is one of the best treatments for the motor symptoms of Parkinson's disease. However, its use is limited by the fact that it provides only symptomatic relief and chronic therapy leads to dyskinesias. There is therefore a continual search for novel therapeutic approaches. Nicotine, a drug that acts at nicotinic acetylcholine receptors (nAChRs), has been shown to protect against nigrostriatal damage and reduce L-dopa-induced dyskinesias. NAChRs may therefore represent novel targets for Parkinson's disease management. Since there are multiple nAChRs throughout the body, it is important to understand the subtypes involved in striatal function to allow for the development of drugs with optimal beneficial effects. Here we discuss recent work from our laboratory which indicates that α6ß2* and α4ß2* nAChRs are key in regulating striatal dopaminergic function. Experiments in parkinsonian rats using cyclic voltammetry showed that both α6ß2* and α4ß2* nAChR-mediated evoked-dopamine release in striatal slices is affected by nigrostriatal damage. These subtypes also appear to be important for neuroprotection against nigrostriatal damage and the nicotine-mediated reduction in L-dopa-induced dyskinesias in parkinsonian animal models. Our combined findings indicate that α4ß2* and α6ß2* nAChRs may represent useful therapeutic targets for Parkinson's disease.

17.
Mol Pharmacol ; 78(5): 971-80, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20732972

ABSTRACT

Nicotinic receptors (nAChRs) are important modulators of dopaminergic transmission in striatum, a region critical to Parkinson's disease. The nAChRs mainly involved are the α6ß2* and α4ß2* subtypes. Lesion studies show that the α6ß2* receptor is decreased to a much greater extent with nigrostriatal damage than the α4ß2* subtype raising the question whether this latter nAChR population is more important with increased nigrostriatal damage. To address this, we investigated the effect of varying nigrostriatal damage on α6ß2* and α4ß2* receptor-modulated dopamine signaling using cyclic voltammetry. This approach offers the advantage that changes in dopamine release can be observed under different neuronal firing conditions. Total single-pulse-evoked dopamine release decreased in direct proportion to declines in the dopamine transporter and dopamine uptake. We next used α-conotoxinMII and mecamylamine to understand the role of the α4ß2* and α6ß2* subtypes in release. Single-pulse-stimulated α6ß2* and α4ß2* receptor dopamine release decreased to a similar extent with increasing nigrostriatal damage, indicating that both subtypes contribute to the control of dopaminergic transmission with lesioning. Total burst-stimulated dopamine release also decreased proportionately with nigrostriatal damage. However, the role of the α4ß2* and α6ß2* nAChRs varied with different degrees of lesioning, suggesting that the two subtypes play a unique function with burst firing, with a somewhat more prominent and possibly more selective role for the α6ß2* subtype. These data have important therapeutic implications because they suggest that drugs directed to both α4ß2* and α6ß2* nAChRs may be useful in the treatment of neurological disorders such as Parkinson's disease.


Subject(s)
Corpus Striatum/metabolism , Dopamine/metabolism , Receptors, Nicotinic/physiology , Substantia Nigra/metabolism , Action Potentials , Animals , Autoradiography , Binding Sites , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Dopamine Plasma Membrane Transport Proteins/metabolism , Male , Neurons/pathology , Neurons/physiology , Nicotinic Antagonists/pharmacology , Oxidopamine , Parkinson Disease/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Substantia Nigra/pathology , Substantia Nigra/physiopathology
18.
Biochem Pharmacol ; 78(7): 677-85, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19433069

ABSTRACT

There exists a remarkable diversity of neurotransmitter compounds in the striatum, a pivotal brain region in the pathology of Parkinson's disease, a movement disorder characterized by rigidity, tremor and bradykinesia. The striatal dopaminergic system, which is particularly vulnerable to neurodegeneration in this disorder, appears to be the major contributor to these motor problems. However, numerous other neurotransmitter systems in the striatum most likely also play a significant role, including the nicotinic cholinergic system. Indeed, there is an extensive anatomical overlap between dopaminergic and cholinergic neurons, and acetylcholine is well known to modulate striatal dopamine release both in vitro and in vivo. Nicotine, a drug that stimulates nicotinic acetylcholine receptors (nAChRs), influences several functions relevant to Parkinson's disease. Extensive studies in parkinsonian animals show that nicotine protects against nigrostriatal damage, findings that may explain the well-established decline in Parkinson's disease incidence with tobacco use. In addition, recent work shows that nicotine reduces l-dopa-induced abnormal involuntary movements, a debilitating complication of l-dopa therapy for Parkinson's disease. These combined observations suggest that nAChR stimulation may represent a useful treatment strategy for Parkinson's disease for neuroprotection and symptomatic treatment. Importantly, only selective nAChR subtypes are present in the striatum including the alpha4beta2*, alpha6beta2* and alpha7 nAChR populations. Treatment with nAChR ligands directed to these subtypes may thus yield optimal therapeutic benefit for Parkinson's disease, with a minimum of adverse side effects.


Subject(s)
Neuroprotective Agents/pharmacology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Parkinson Disease/metabolism , Animals , Antiparkinson Agents/adverse effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Humans , Levodopa/adverse effects , Motor Skills/drug effects , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Receptors, Nicotinic/physiology , Smoking/adverse effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/physiopathology
19.
Mol Pharmacol ; 75(4): 938-46, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19144785

ABSTRACT

Brain dopaminergic systems are critical in motor control as evidenced by findings that their disruption results in movement disorders such as Parkinson's disease. Nicotinic acetylcholine receptor (nAChR) activation plays an important role in regulating striatal dopaminergic function. Rodent studies show that short-term nicotine exposure influences stimulated striatal dopamine release with responsiveness dependent on neuronal activity. However, studies have not yet been done in nonhuman primates, nor has work been done to evaluate the effect of long-term nicotine exposure, which is relevant for therapies for chronic neurological disorders. Here, we used voltammetry to assess the role of nAChRs on evoked dopamine release from monkey putamen slices. In both ventral and dorsal putamen, alpha3/alpha6beta2(*) nAChRs regulated > or =80% of non-burst- (single pulse) nAChR-modulated dopamine release, and alpha4beta2(*) nAChRs regulated the remainder. Similar results were observed with burst-firing in ventral but not dorsal putamen, indicating that nAChR-modulated effects on release depend on the subregion and firing frequency. Next, we investigated the consequence of long-term nicotine exposure via the drinking water on nAChR-modulated responsiveness. Nicotine treatment altered both non-burst- and burst-stimulated dopamine release in ventral but not dorsal putamen. Altogether, these data support a predominant role for alpha3/alpha6beta2(*) nAChRs in the regulation of evoked dopamine release in nonhuman primate putamen. They also show that long-term nicotine treatment selectively modifies nAChR-modulated release in distinct striatal subregions. These findings have implications for the development of treatments for addiction and neurological disorders with nAChR dysfunction.


Subject(s)
Dopamine/metabolism , Nicotine/administration & dosage , Putamen/metabolism , Receptors, Nicotinic/physiology , Animals , Dose-Response Relationship, Drug , Female , Putamen/drug effects , Saimiri , Time Factors
20.
Mol Pharmacol ; 74(3): 844-53, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18583454

ABSTRACT

Nicotine treatment has long been associated with alterations in alpha4beta2(*) nicotinic acetylcholine receptor (nAChR) expression that modify dopaminergic function. However, the influence of long-term nicotine treatment on the alpha6beta2(*) nAChR, a subtype specifically localized on dopaminergic neurons, is less clear. Here we used voltammetry, as well as receptor binding studies, to identify the effects of nicotine on striatal alpha6beta2(*) nAChR function and expression. Long-term nicotine treatment via drinking water enhanced nonburst and burst endogenous dopamine release from rat striatal slices. In control animals, alpha6beta2(*) nAChR blockade with alpha-conotoxin MII (alpha-CtxMII) decreased release with nonburst stimulation but not with burst firing. These data in control animals suggest that varying stimulus frequencies differentially regulate alpha6beta2(*) nAChR-evoked dopamine release. In contrast, in nicotine-treated rats, alpha6beta2(*) nAChR blockade elicited a similar pattern of dopamine release with nonburst and burst firing. To elucidate the alpha6beta2(*) nAChR subtypes altered with long-term nicotine treatment, we used the novel alpha-CtxMII analog E11A in combination with alpha4 nAChR knockout mice. (125)I-alpha-CtxMII competition studies in striatum of knockout mice showed that nicotine treatment decreased the alpha6alpha4beta2(*) subtype but increased the alpha6(nonalpha4)beta2(*) nAChR population. These data indicate that alpha6beta2(*) nAChR-evoked dopamine release in nicotine-treated rats is mediated by the alpha6(nonalpha4)beta2(*) nAChR subtype and suggest that the alpha6alpha4beta2(*) nAChR and/or alpha4beta2(*) nAChR contribute to the differential effect of higher frequency stimulation on dopamine release under control conditions. Thus, alpha6beta2(*) nAChR subtypes may represent important targets for smoking cessation therapies and neurological disorders involving these receptors such as Parkinson's disease.


Subject(s)
Gene Expression Regulation/drug effects , Neostriatum/metabolism , Nicotine/administration & dosage , Nicotine/pharmacology , Receptors, Nicotinic/metabolism , Action Potentials/drug effects , Animals , Dopamine/metabolism , Down-Regulation/drug effects , Electrophysiology , Male , Mice , Neostriatum/drug effects , Rats , Rats, Sprague-Dawley , Time Factors , Water
SELECTION OF CITATIONS
SEARCH DETAIL
...