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










Publication year range
1.
Neuropharmacology ; 235: 109547, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37116611

ABSTRACT

Nicotine is the principal psychoactive component in tobacco that drives addiction through its action on neuronal nicotinic acetylcholine receptors (nAChR). The nicotinic receptor gene CHRNA5, which encodes the α5 subunit, is associated with nicotine use and dependence. In humans, the CHRNA5 missense variant rs16969968 (G > A) is associated with increased risk for nicotine dependence and other smoking-related phenotypes. In rodents, α5-containing nAChRs in dopamine (DA) neurons within the ventral tegmental area (VTA) powerfully modulate nicotine reward and reinforcement. Although the neuroadaptations caused by long-term nicotine exposure are being actively delineated at both the synaptic and behavioral levels, the contribution of α5-containing nAChRs to the cellular adaptations associated with long-term nicotine exposure remain largely unknown. To gain insight into the mechanisms behind the influence of α5-containing nAChRs and the rs16969968 polymorphism on nicotine use and dependence, we used electrophysiological approaches to examine changes in nAChR function arising in VTA neurons during chronic nicotine exposure and multiple stages of nicotine withdrawal. Our results demonstrate that CHRNA5 mutation leads to profound changes in VTA nAChR function at baseline, during chronic nicotine exposure, and during short-term and prolonged withdrawal. Whereas nAChR function was suppressed in DA neurons from WT mice undergoing withdrawal relative to drug-naïve or nicotine-drinking mice, α5-null mice exhibited an increase in nAChR function during nicotine exposure that persisted throughout 5-10 weeks of withdrawal. Re-expressing the hypofunctional rs16969968 CHRNA5 variant in α5-null VTA DA neurons did not rescue the phenotype, with α5-SNP neurons displaying a similar increased response to ACh during nicotine exposure and early stages of withdrawal. These results demonstrate the importance of VTA α5-nAChRs in the response to nicotine and implicate them in the time course of withdrawal.


Subject(s)
Nicotine , Receptors, Nicotinic , Humans , Mice , Animals , Nicotine/pharmacology , Dopaminergic Neurons/metabolism , Ventral Tegmental Area/metabolism , Receptors, Nicotinic/metabolism , Smoking , Mice, Knockout , Nerve Tissue Proteins/genetics
2.
J Diabetes Res ; 2022: 7083106, 2022.
Article in English | MEDLINE | ID: mdl-35915720

ABSTRACT

Studies on nutrient sequences during meals suggest that consuming carbohydrates last lowers postprandial glucose excursions more than consuming carbohydrates first. However, this phenomenon has not been studied in gestational diabetes mellitus (GDM). Ten women with GDM consumed the same caloric foods in different sequences over five successive days: (A) dish first, followed by carbohydrate and soup last; (B) carbohydrate first, followed by dish and soup last; (C) soup first, followed by dish and carbohydrate last; (D) three meals a day ad libitum; and (E) six meals a day as ad libitum. Continuous glucose monitoring (CGM) was used to assess diurnal glycemia. Decreases in mean glucose levels and the largest glucose levels in A were similar to group C. The peak glucose of breakfast and lunch in group B was more significant than in groups A and C. The B meal pattern showed more marked glycemic excursions than groups A and C. Increasing the number of meals reduced the peak glucose level and the glycemic excursions with the same total calories. Changing meal sequences or increasing the number of meals may reduce glycemic excursions in GDM. Our trial was registered retrospectively and the trial registration number is ChiCTR2200057044.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetes, Gestational , Blood Glucose , Blood Glucose Self-Monitoring , Dietary Carbohydrates , Female , Glucose , Humans , Insulin , Meals , Postprandial Period , Pregnancy , Retrospective Studies
3.
Neuropharmacology ; 218: 109218, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35973602

ABSTRACT

The single nucleotide polymorphism (SNP) D398N (rs16969968) in CHRNA5, the gene encoding the α5 subunit of the nicotinic acetylcholine receptors (nAChR), has been associated with both nicotine and opiate dependence in human populations. Expression of this SNP on presynaptic VTA dopaminergic (DA) neurons is known to cause a reduction in calcium signaling, leading to alterations in transmitter signaling and altered responses to drugs of abuse. To examine the impact of the Chrna5 SNP on opiate reward and underlying dopaminergic mechanisms, mice harboring two copies of the risk-associated allele (Chrna5 A/A) at a location equivalent to human rs16969968 were generated via CRISPR/cas9 genome editing. We sought to determine whether Chrna5 A/A mice show differences in sensitivity to rewarding properties of morphine using the conditioned place preference paradigm. When mice were tested two weeks after conditioning, female Chrna5 A/A mice showed significantly enhanced preference for the morphine-paired chamber relative to WT females, suggesting that this genotype may enhance opioid reward specifically in females. In contrast, Chrna5 genotype had no effect on locomotor sensitization in male or female mice. Relative to WT females, peak amplitude of ACh-gated currents recorded from VTA DA neurons in Chrna5 A/A females was potentiated 1 day after conditioning with morphine. Increased FOS expression was also observed in Chrna5 A/A mice relative to WT mice following exposure to the morphine CPP chamber. We propose that impaired α5 nAChR subunit function alters DA neuron response following repeated morphine exposures, and that this early cellular response could contribute to enhanced opiate reward two weeks after conditioning.


Subject(s)
Polymorphism, Single Nucleotide , Receptors, Nicotinic , Animals , Female , Male , Mice , Morphine/pharmacology , Nerve Tissue Proteins/metabolism , Nicotine/pharmacology , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Reward
4.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Article in English | MEDLINE | ID: mdl-34580198

ABSTRACT

Aversive memories are important for survival, and dopaminergic signaling in the hippocampus has been implicated in aversive learning. However, the source and mode of action of hippocampal dopamine remain controversial. Here, we utilize anterograde and retrograde viral tracing methods to label midbrain dopaminergic projections to the dorsal hippocampus. We identify a population of midbrain dopaminergic neurons near the border of the substantia nigra pars compacta and the lateral ventral tegmental area that sends direct projections to the dorsal hippocampus. Using optogenetic manipulations and mutant mice to control dopamine transmission in the hippocampus, we show that midbrain dopamine potently modulates aversive memory formation during encoding of contextual fear. Moreover, we demonstrate that dopaminergic transmission in the dorsal CA1 is required for the acquisition of contextual fear memories, and that this acquisition is sustained in the absence of catecholamine release from noradrenergic terminals. Our findings identify a cluster of midbrain dopamine neurons that innervate the hippocampus and show that the midbrain dopamine neuromodulation in the dorsal hippocampus is sufficient to maintain aversive memory formation.


Subject(s)
Dopamine/metabolism , Hippocampus/metabolism , Memory/physiology , Animals , Dopaminergic Neurons , Fear/physiology , Female , Learning/physiology , Male , Mesencephalon/physiology , Mice , Mice, Inbred C57BL , Optogenetics/methods , Substantia Nigra/metabolism , Ventral Tegmental Area/physiology
5.
Exp Ther Med ; 22(4): 1071, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34447464

ABSTRACT

Accumulating researches indicate that long non-coding RNAs (lncRNAs) participate in human bone mesenchymal stem cells (hBMSCs) osteogenic differentiation. The present study aimed to investigate the underlying molecular mechanisms of long intergenic non-protein coding RNA 899 (LINC00899) in osteoporosis. Therefore, reverse transcription-quantitative PCR was performed to evaluate the expression levels of LINC00899, microRNA (miR)-374a and runt-related transcription factor 2 (RUNX2) in clinical tissues and hBMSCs. The potential interaction between miR-374a and LINC00899 or RUNX2 was predicted utilizing the StarBase software and verified by luciferase reporter and RNA binding protein immunoprecipitation assays. In addition, alkaline phosphatase activity and Alizarin Red S staining were used to evaluate the osteogenic potential of hBMSCs. The results showed that the expression levels of LINC00899 were gradually increased, whilst those of miR-374a were decreased as the osteogenic induction process progresses. Additionally, the expression of LINC00899 was downregulated in the bone tissues of patients with osteoporosis, where LINC00899 knockdown reduced the expression levels of osteogenesis-related genes osteocalcin (OCN), osteopontin (OPN) and RUNX2 in hBMSCs. LINC00899 was also found to directly target miR-374a, thereby inhibiting its expression. Finally, it was predicted that RUNX2 could be directly targeted by miR-374a, such that miR-374a silencing partially abolished the inhibitory effect of LINC00899 knockdown on the expression of RUNX2, OPN and OCN. Overall, findings of the present study suggested that LINC00899 could facilitate the osteogenic differentiation of hBMSCs and prevent osteoporosis by sponging miR-374a to enhance the expression of RUNX2, which provide a potentially useful therapeutic strategy for patients with osteoporosis.

6.
Inflammation ; 44(3): 965-973, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33566259

ABSTRACT

Long noncoding RNAs (lncRNAs) have been reported to be implicated in various biological and pathological processes. However, the function and mechanism of XIST in vascular smooth muscle cells (VSMCs) remains unknown. The levels of XIST, miR-599, and TLR4 were tested by RT-qPCR. VSMCs and human mononuclear cells (U937) treated with ox-LDL were used as atherosclerosis (AS) cell models. The CCK-8 assay was adopted to detect cell viability. Cell apoptosis was examined by the TUNEL assay. A dual-luciferase reporter assay was employed to investigate the interaction between miR-599 and XIST or TLR4. In this research, we uncovered that the XIST level was elevated in the serum of AS patients and ox-LDL-treated AS cell models. Functional analysis revealed that XIST depletion restrained cell proliferation, while induced the apoptosis in AS cell models. Besides, miR-599 was verified to be a direct downstream target of XIST and miR-599 inhibitor reversed the effects of XIST knockdown on AS progression. Finally, we demonstrated that XIST increased TLR4 expression by serving as a ceRNA of miR-599. All these findings manifested the role of the XIST/miR-599/TLR4 axis in AS development.


Subject(s)
Atherosclerosis/metabolism , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , RNA, Long Noncoding/metabolism , Toll-Like Receptor 4/metabolism , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/pathology , Case-Control Studies , Cell Proliferation , Disease Progression , Humans , Lipoproteins, LDL/toxicity , MicroRNAs/genetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , RNA, Long Noncoding/genetics , Signal Transduction , Toll-Like Receptor 4/genetics , U937 Cells
7.
Eur J Neurosci ; 45(1): 138-146, 2017 01.
Article in English | MEDLINE | ID: mdl-27646422

ABSTRACT

Physiological and behavioral evidence supports that dopamine (DA) receptor signaling influences hippocampal function. While several recent studies examined how DA influences CA1 plasticity and learning, there are fewer studies investigating the influence of DA signaling to the dentate gyrus. The dentate gyrus receives convergent cortical input through the perforant path fiber tracts and has been conceptualized to detect novelty in spatial memory tasks. To test whether DA-receptor activity influences novelty-detection, we used a novel object recognition (NOR) task where mice remember previously presented objects as an indication of learning. Although DA innervation arises from other sources and the main DA signaling may be from those sources, our molecular approaches verified that midbrain dopaminergic fibers also sparsely innervate the dentate gyrus. During the NOR task, wild-type mice spent significantly more time investigating novel objects rather than previously observed objects. Dentate granule cells in slices cut from those mice showed an increased AMPA/NMDA-receptor current ratio indicative of potentiated synaptic transmission. Post-training injection of a D1-like receptor antagonist not only effectively blocked the preference for the novel objects, but also prevented the increased AMPA/NMDA ratio. Consistent with that finding, neither NOR learning nor the increase in the AMPA/NMDA ratio were observed in DA-receptor KO mice under the same experimental conditions. The results indicate that DA-receptor signaling contributes to the successful completion of the NOR task and to the associated synaptic plasticity of the dentate gyrus that likely contributes to the learning.


Subject(s)
Hippocampus/physiology , Neuronal Plasticity/physiology , Receptors, Dopamine/metabolism , Recognition, Psychology/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Dopamine/metabolism , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Mice, Knockout , Neuronal Plasticity/genetics , Receptors, Dopamine/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
8.
Cell Rep ; 14(8): 1930-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26904943

ABSTRACT

Dopamine release during reward-driven behaviors influences synaptic plasticity. However, dopamine innervation and release in the hippocampus and its role during aversive behaviors are controversial. Here, we show that in vivo hippocampal synaptic plasticity in the CA3-CA1 circuit underlies contextual learning during inhibitory avoidance (IA) training. Immunohistochemistry and molecular techniques verified sparse dopaminergic innervation of the hippocampus from the midbrain. The long-term synaptic potentiation (LTP) underlying the learning of IA was assessed with a D1-like dopamine receptor agonist or antagonist in ex vivo hippocampal slices and in vivo in freely moving mice. Inhibition of D1-like dopamine receptors impaired memory of the IA task and prevented the training-induced enhancement of both ex vivo and in vivo LTP induction. The results indicate that dopamine-receptor signaling during an aversive contextual task regulates aversive memory retention and regulates associated synaptic mechanisms in the hippocampus that likely underlie learning.


Subject(s)
Avoidance Learning/physiology , CA1 Region, Hippocampal/physiology , Dopamine/metabolism , Learning/physiology , Long-Term Potentiation/physiology , Memory, Long-Term/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Avoidance Learning/drug effects , Benzazepines/pharmacology , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/cytology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Electrodes , Long-Term Potentiation/drug effects , Memory, Long-Term/drug effects , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/physiology , Mice , Mice, Inbred C57BL , Microtomy , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/physiology , Synapses/drug effects , Synapses/physiology , Synapses/ultrastructure , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tissue Culture Techniques
9.
Neuropharmacology ; 90: 23-32, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25445492

ABSTRACT

Attention-deficit hyperactive disorder (ADHD) is the most commonly studied and diagnosed psychiatric disorder in children. Methylphenidate (MPH, e.g., Ritalin) has been used to treat ADHD for over 50 years. It is the most commonly prescribed treatment for ADHD, and in the past decade it was the drug most commonly prescribed to teenagers. In addition, MPH has become one of the most widely abused drugs on college campuses. In this study, we examined the effects of MPH on hippocampal synaptic plasticity, which serves as a measurable quantification of memory mechanisms. Field potentials were recorded with permanently implanted electrodes in freely-moving mice to quantify MPH modulation of perforant path synaptic transmission onto granule cells of the dentate gyrus. Our hypothesis was that MPH affects hippocampal synaptic plasticity underlying learning because MPH boosts catecholamine signaling by blocking the dopamine and norepinephrine transporters (DAT and NET respectively). In vitro hippocampal slice experiments indicated MPH enhances perforant path plasticity, and this MPH enhancement arose from action via D1-type dopamine receptors and ß-type adrenergic receptors. Similarly, MPH boosted in vivo initiation of long-term potentiation (LTP). While there was an effect via both dopamine and adrenergic receptors in vivo, LTP induction was more dependent on the MPH-induced action via D1-type dopamine receptors. Under biologically reasonable experimental conditions, MPH enhances hippocampal synaptic plasticity via catecholamine receptors.


Subject(s)
Central Nervous System Stimulants/pharmacology , Dentate Gyrus/drug effects , Long-Term Potentiation/drug effects , Methylphenidate/pharmacology , Receptors, Adrenergic/metabolism , Receptors, Dopamine/metabolism , Animals , Dentate Gyrus/physiology , Dopamine/metabolism , Dose-Response Relationship, Drug , Electric Stimulation , Electrodes, Implanted , Female , Long-Term Potentiation/physiology , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Perforant Pathway/drug effects , Perforant Pathway/physiology , Theta Rhythm/physiology , Tissue Culture Techniques
10.
J Neurosci ; 34(48): 15888-97, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25429131

ABSTRACT

Although evidence suggests that DA modulates hippocampal function, the mechanisms underlying that dopaminergic modulation are largely unknown. Using perforated-patch electrophysiological techniques to maintain the intracellular milieu, we investigated how the activation of D1-type DA receptors regulates spike timing-dependent plasticity (STDP) of the medial perforant path (mPP) synapse onto dentate granule cells. When D1-type receptors were inhibited, a relatively mild STDP protocol induced LTP only within a very narrow timing window between presynaptic stimulation and postsynaptic response. The stimulus protocol produced timing-dependent LTP (tLTP) only when the presynaptic stimulation was followed 30 ms later by depolarization-induced postsynaptic action potentials. That is, the time between presynaptic stimulation and postsynaptic response was 30 ms (Δt = +30 ms). When D1-type receptors were activated, however, the same mild STDP protocol induced tLTP over a much broader timing window: tLTP was induced when -30 ms ≤ Δt ≤ +30 ms. The result indicated that D1-type receptor activation enabled synaptic potentiation even when postsynaptic activity preceded presynaptic stimulation within this Δt range. Results with null mice lacking the Kv4.2 potassium channel and with the potassium channel inhibitor, 4-aminopyridine, suggested that D1-type receptors enhanced tLTP induction by suppressing the transient IA-type K(+) current. Results obtained with antagonists and DA receptor knock-out mice indicated that endogenous activity of both D1 and D5 receptors modulated plasticity in the mPP. The DA D5 receptors appeared particularly important in regulating plasticity of the mPP onto the dentate granule cells.


Subject(s)
Dentate Gyrus/physiology , Neuronal Plasticity/physiology , Perforant Pathway/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D5/physiology , Synapses/physiology , Action Potentials/physiology , Animals , Excitatory Postsynaptic Potentials/physiology , Female , Long-Term Potentiation/physiology , Male , Mice , Mice, Knockout , Organ Culture Techniques , Time Factors
11.
Ann Neurol ; 69(4): 664-72, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21391233

ABSTRACT

OBJECTIVE: Human hypothalamic hamartomas (HHs) are highly associated with treatment-resistant gelastic seizures. HHs are intrinsically epileptogenic, although the basic cellular mechanisms responsible for seizure activity are unknown. Altered gamma-aminobutyric acid (GABA) function can contribute to epileptogenesis in humans and animal models. Recently, functional GABA(A) receptor (GABA(A) R) rundown has been described in surgically resected human temporal lobe epilepsy tissue. We asked whether functional GABA(A) R rundown also occurs in human HH neurons. METHODS: GABA(A) R-mediated currents were measured using perforated patch-clamp recordings in single neurons acutely dissociated from surgically resected HH tissue. In addition, functional GABA(A) Rs were expressed in Xenopus oocytes after microinjection with membrane fractions from either HH or control hypothalamus, and were studied with 2-electrode voltage-clamp recordings. RESULTS: Perforated patch-clamp recordings in dissociated HH neurons showed that repetitive exposure to GABA (5 consecutive exposures to 0.1 mM GABA with 1-second duration and at 20-second intervals) induced a time-dependent rundown of whole-cell currents in small HH neurons, whereas large HH neurons showed much less rundown using the same protocol. Functional rundown was not observed in HH neurons with repetitive exposure to glycine or glutamate. Two-electrode voltage-clamp recordings (6 consecutive exposures to 1 mM GABA with 10-second duration and at 40-second intervals) induced GABA current rundown in Xenopus oocytes microinjected with HH membrane proteins, but not in the oocytes expressing hypothalamic membrane proteins derived from human autopsy controls. Functional rundown of GABA currents was significantly attenuated by intracellular application of adenosine triphosphate or the nonspecific phosphatase inhibitor, okadaic acid. INTERPRETATION: Neurons from surgically resected human HH demonstrate functional rundown of GABA(A) R-mediated transmembrane currents in response to GABA agonist exposure. Rundown may be a marker for impaired GABAergic function and a contributing mechanism for seizure genesis within HH tissue.


Subject(s)
Epilepsy/metabolism , Neurons/metabolism , Receptors, GABA-A/metabolism , Animals , Epilepsy/etiology , Hamartoma/complications , Hamartoma/metabolism , Hamartoma/pathology , Humans , Hypothalamic Diseases/complications , Hypothalamic Diseases/metabolism , Hypothalamic Diseases/pathology , Hypothalamus/metabolism , Oocytes , Patch-Clamp Techniques/instrumentation , Patch-Clamp Techniques/methods , Xenopus
12.
J Neurosci ; 31(7): 2537-48, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21325521

ABSTRACT

Diverse nicotinic acetylcholine receptor (nAChR) subtypes containing different subunit combinations can be placed on nerve terminals or soma/dendrites in the ventral tegmental area (VTA). nAChR α6 subunit message is abundant in the VTA, but α6*-nAChR cellular localization, function, pharmacology, and roles in cholinergic modulation of dopaminergic (DA) neurons within the VTA are not well understood. Here, we report evidence for α6ß2*-nAChR expression on GABA neuronal boutons terminating on VTA DA neurons. α-Conotoxin (α-Ctx) MII labeling coupled with immunocytochemical staining localizes putative α6*-nAChRs to presynaptic GABAergic boutons on acutely dissociated, rat VTA DA neurons. Functionally, acetylcholine (ACh) induces increases in the frequency of bicuculline-, picrotoxin-, and 4-aminopyridine-sensitive miniature IPSCs (mIPSCs) mediated by GABA(A) receptors. These increases are abolished by α6*-nAChR-selective α-Ctx MII or α-Ctx PIA (1 nm) but not by α7 (10 nm methyllycaconitine) or α4* (1 µm dihydro-ß-erythroidine)-nAChR-selective antagonists. ACh also fails to increase mIPSC frequency in VTA DA neurons prepared from nAChR ß2 knock-out mice. Moreover, ACh induces an α-Ctx PIA-sensitive elevation in intraterminal Ca(2+) in synaptosomes prepared from the rat VTA. Subchronic exposure to 500 nm nicotine reduces ACh-induced GABA release onto the VTA DA neurons, as does 10 d of systemic nicotine exposure. Collectively, these results indicate that α6ß2*-nAChRs are located on presynaptic GABAergic boutons within the VTA and modulate GABA release onto DA neurons. These presynaptic α6ß2*-nAChRs likely play important roles in nicotinic modulation of DA neuronal activity.


Subject(s)
Dopamine/metabolism , Neurons/cytology , Presynaptic Terminals/physiology , Receptors, Nicotinic/metabolism , Ventral Tegmental Area/cytology , gamma-Aminobutyric Acid/metabolism , 4-Aminopyridine/pharmacology , Acetylcholine/pharmacology , Aconitine/analogs & derivatives , Aconitine/pharmacology , Animals , Animals, Newborn , Bungarotoxins/pharmacology , Calcium/metabolism , Conotoxins/metabolism , Dihydro-beta-Erythroidine/pharmacology , Dopamine/pharmacology , Drug Interactions , Excitatory Amino Acid Agents/pharmacology , Female , GABA Agents/pharmacology , Glutamate Decarboxylase/metabolism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice , Mice, Knockout , Neurons/drug effects , Nicotine/pharmacology , Nicotinic Antagonists/pharmacology , Potassium Channel Blockers/pharmacology , Presynaptic Terminals/drug effects , Protein Binding/drug effects , Rats , Rats, Wistar , Receptors, Nicotinic/deficiency , Synaptosomes/drug effects , Synaptosomes/metabolism
13.
Synapse ; 65(4): 332-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20730803

ABSTRACT

Nicotine promotes glutamatergic synaptic plasticity in dopaminergic (DA) neurons in the ventral tegmental area (VTA), which is thought to be an important mechanism underlying nicotine reward. However, it is unclear whether exposure of nicotine alone to VTA slice is sufficient to increase glutamatergic synaptic strength on DA neurons and which nicotinic acetylcholine receptor (nAChR) subtype mediates this effect. Here, we report that the incubation of rat VTA slices with 500 nM nicotine induces glutamatergic synaptic plasticity in DA neurons. We measure the ratio of AMPA and NMDA receptor-mediated currents (AMPA/NMDA) and compare these ratios between nicotine-treated and -untreated slices. Our results demonstrate that the incubation of VTA slices with 500 nM nicotine for 1 h (but not for 10 min) significantly increases the AMPA/NMDA ratio when compared with controls. Preincubation with 10 nM of the α7-nAChR antagonist, methyllycaconitine (MLA) but not 1 µM α4-containing nAChR antagonist, dihydro-ß-erythroidine (DHßE) prevents nicotinic effect, suggesting that α7-nAChRs are mainly mediated this nicotinic effect. This finding is further supported by the disappearance of this nicotinic effect in nAChR α7 knockout (KO) mice. Furthermore, nicotine reduced paired-pulse ratio (PPR) of evoked excitatory postsynaptic potential (eEPSP) in the VTA slices prepared from wild-type (WT) mice but not α7 KO mice. Collectively, these findings suggest that exposure of smoking-relevant concentrations of nicotine to VTA slices is sufficient to increase glutamatergic synaptic strength on DA neurons and that α7-nAChRs likely mediate this nicotinic effect through increasing presynaptic release of glutamate.


Subject(s)
Dopamine/metabolism , Glutamic Acid/metabolism , Neuronal Plasticity/physiology , Nicotine/administration & dosage , Synapses/metabolism , Ventral Tegmental Area/metabolism , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Synapses/drug effects , Time Factors , Ventral Tegmental Area/drug effects
14.
J Neurosci ; 30(41): 13814-25, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-20943922

ABSTRACT

Systemic exposure to nicotine induces glutamatergic synaptic plasticity on dopamine (DA) neurons in the ventral tegmental area (VTA), but mechanisms are largely unknown. Here, we report that single, systemic exposure in rats to nicotine (0.17 mg/kg free base) increases the ratio of DA neuronal currents mediated by AMPA relative to NMDA receptors (AMPA/NMDA ratio) assessed 24 h later, based on slice-patch recording. The AMPA/NMDA ratio increase is evident within 1 h and lasts for at least 72 h after nicotine exposure (and up to 8 d after repeated nicotine administration). This effect cannot be prevented by systemic injection of either α7-nAChR (nicotinic ACh receptor)-selective [methyllycaconitine (MLA)] or ß2*-nAChR-selective [mecamylamine (MEC)] antagonists but is prevented by coinjection of MLA and MEC. In either nAChR α7 or ß2 subunit knock-out mice, systemic exposure to nicotine still increases the AMPA/NMDA ratio. Preinjection in rats of a NMDA receptor antagonist MK-801((+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine maleate), but neither DA receptor antagonists [SCH-23390 (R-(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine) plus haloperidol] nor a calcineurin inhibitor (cyclosporine), prevents the nicotine-induced increase in AMPA/NMDA ratio. After systemic exposure to nicotine, glutamatergic (but not GABAergic) transmission onto rat VTA DA neuronal inputs is enhanced. Correspondingly, DA neuronal firing measured 24 h after nicotine exposure using extracellular single-unit recording in vivo is significantly faster, and there is conversion of silent to active DA neurons. Collectively, these findings demonstrate that systemic nicotine acting via either α7- or ß2*-nAChRs increases presynaptic and postsynaptic glutamatergic function, and consequently initiates glutamatergic synaptic plasticity, which may be an important, early neuronal adaptation in nicotine reward and reinforcement.


Subject(s)
Dopamine/metabolism , Glutamic Acid/metabolism , Neuronal Plasticity/physiology , Neurons/physiology , Nicotine/metabolism , Synapses/physiology , Ventral Tegmental Area/physiology , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Genotype , Mice , Mice, Knockout , Neuronal Plasticity/drug effects , Neurons/drug effects , Nicotine/pharmacology , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synapses/drug effects , Ventral Tegmental Area/drug effects , alpha7 Nicotinic Acetylcholine Receptor
15.
Neuropharmacology ; 59(7-8): 567-72, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20804776

ABSTRACT

Tetrahydroberberine (THB) exhibits neuroprotective effects but its targets and underlying mechanisms are largely unknown. Emerging evidence indicates that ATP-sensitive potassium (K(ATP)) channels in the substantia nigra pars compacta (SNc) promote Parkinson disease (PD) pathogenesis, thus blocking K(ATP) channels may protect neurons against neuronal degeneration. In the present study, we tested a hypothesis that THB blocks K(ATP) channels in dopaminergic (DA) neurons acutely dissociated from rat SNc. Using perforated patch-clamp recording in current-clamp mode, the functional K(ATP) channels can be opened by persistent perfusion of rotenone, an inhibitor of complex I of the mitochondrial respiratory chain. Bath-application of THB reversibly blocks opened K(ATP) channels in a concentration-dependent manner, which is comparable to a classical K(ATP) channel blocker, Tol. Compared to THB analogs, l-stepholidine (l-SPD) or l-tetrahydropalmatine (l-THP), THB exhibits more profound blockade in K(ATP) channels. In addition, exposure of THB alone to the recorded neuron increases action potential firing, and THB also restores rotenone-induced membrane hyperpolarization in the presence of dopamine D2 receptor antagonist (sulpiride), suggesting that THB exhibits an excitatory effect on SNc DA neurons through the block of K(ATP) channels. Collectively, the blockade of neuronal K(ATP) channels by THB in SNc DA neurons is a novel pharmacological mechanism of THB, which may contribute to its neuroprotective effects in PD.


Subject(s)
Berberine/analogs & derivatives , Dopamine/metabolism , KATP Channels/antagonists & inhibitors , Neurons/drug effects , Neuroprotective Agents/pharmacology , Substantia Nigra/drug effects , Animals , Berberine/pharmacology , In Vitro Techniques , Neurons/physiology , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, Dopamine D2/physiology , Substantia Nigra/cytology , Substantia Nigra/physiology
16.
J Pharmacol Exp Ther ; 335(2): 401-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20688974

ABSTRACT

Lamotrigine (LTG), an anticonvulsive drug, is often used for the treatment of a variety of epilepsies. In addition to block of sodium channels, LTG may act on other targets to exert its antiepileptic effect. In the present study, we evaluated the effects of LTG on neuronal nicotinic acetylcholine receptors (nAChRs) using the patch-clamp technique on human α4ß2-nAChRs heterologously expressed in the SH-EP1 cell line and on native α4ß2-nAChRs in dopaminergic (DA) neurons in rat ventral tegmental area (VTA). In SH-EP1 cells, LTG diminished the peak and steady-state components of the inward α4ß2-nAChR-mediated currents. This effect exhibited concentration-, voltage- and use-dependent behavior. Nicotine dose-response curves showed that in the presence of LTG, the nicotine-induced maximal current was reduced, suggesting a noncompetitive inhibition. These findings suggest that LTG inhibits human neuronal α4ß2-nAChR function through an open-channel blocking mechanism. LTG-induced inhibition in α4ß2-nAChRs was more profound when preceded by a 2-min pretreatment, after which the nicotine-induced current was reduced even without coapplication of LTG, suggesting that LTG is also able to inhibit α4ß2-nAChRs without channel activation. In freshly dissociated VTA DA neurons, LTG inhibited α4ß2-nAChR-mediated currents but did not affect glutamate- or GABA-induced currents, indicating that LTG selectively inhibits nAChR function. Collectively, our data suggest that the neuronal α4ß2-nAChR is likely an important target for mediating the anticonvulsive effect of LTG and the blockade of α4ß2-nAChR possibly underlying the mechanism through which LTG effectively controls some types of epilepsy, such as autosomal dominant nocturnal frontal lobe epilepsy or juvenile myoclonic epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Neurons/drug effects , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/metabolism , Triazines/pharmacology , Action Potentials/drug effects , Animals , Cell Line , Dopamine/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lamotrigine , Neurons/metabolism , Patch-Clamp Techniques , Rats , Receptors, Nicotinic/genetics , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
17.
Acta Pharmacol Sin ; 30(6): 740-51, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19498417

ABSTRACT

Neuronal nicotinic acetylcholine receptors (nAChRs) are the superfamily of ligand-gated ion channels and widely expressed throughout the central and peripheral nervous systems. nAChRs play crucial roles in modulating a wide range of higher cognitive functions by mediating presynaptic, postsynaptic, and extrasynaptic signaling. Thus far, nine alpha (alpha2-alpha10) and three beta (beta2, beta3, and beta4) subunits have been identified in the CNS, and these subunits assemble to form a diversity of functional nAChRs. Although alpha4beta2- and alpha7-nAChRs are the two major functional nAChR types in the CNS, alpha6*-nAChRs are abundantly expressed in the midbrain dopaminergic (DAergic) system, including mesocorticolimbic and nigrostriatal pathways, and particularly present in presynaptic nerve terminals. Recently, functional and pharmacological profiles of alpha6*-nAChRs have been assessed with the use of alpha6 subunit blockers such as alpha-conotoxin MII and PIA, and also by using alpha6 subunit knockout mice. By modulating DA release in the nucleus accumbens (NAc) and modulating GABA release onto DAergic neurons in the ventral tegmental area (VTA), alpha6*-nAChRs may play important roles in the mediation of nicotine reward and addiction. Furthermore, alpha6*-nAChRs in the nigrostriatal DAergic system may be promising targets for selective preventative treatment of Parkinson's disease (PD). Thus, alpha6*-nAChRs may hold promise for future clinical treatment of human disorders, such as nicotine addiction and PD. In this review, we mainly focus on the recent advances in the understanding of alpha6*-nAChR function, pharmacology and pathophysiology.


Subject(s)
Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/metabolism , Animals , Dopamine/metabolism , Drug Delivery Systems , Humans , Nicotine/adverse effects , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Protein Subunits , Receptors, Nicotinic/drug effects , Reward , Tobacco Use Disorder/physiopathology , Tobacco Use Disorder/rehabilitation , Ventral Tegmental Area/metabolism
18.
Acta Pharmacol Sin ; 30(6): 851-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19498424

ABSTRACT

AIM: Dopaminergic neurons in the substantia nigra pars compacta (SNc) play important roles in motor control and drug addiction. As the major afferent, GABAergic innervation controls the activity of SNc dopaminergic neurons. Although it is clear that nicotine modulates SNc dopaminergic neurons by activating subtypes of somatodendritic nicotinic acetylcholine receptors (nAChRs), the detailed mechanisms of this activation remain to be addressed. METHODS: In the current study, we recorded GABA(A) receptor-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) from dissociated SNc dopaminergic neurons that were obtained using an enzyme-free procedure. These neurons preserved some functional terminals after isolation, including those that release GABA. RESULTS: We found that both extra- and intra-cellular calcium modulates sIPSCs in these neurons. Furthermore, both nicotine and endogenous acetylcholine enhance the frequency of sIPSCs. Moreover, endogenous acetylcholine tonically facilitates sIPSC frequency, primarily by activating the alpha4beta2* nAChRs on the GABAergic terminals. CONCLUSION: Nicotine facilitates GABA release onto SNc dopaminergic neurons mainly via the activation of presynaptic alpha4beta2* nAChRs.


Subject(s)
Dopamine/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, GABA-A/metabolism , Acetylcholine/metabolism , Animals , Calcium/metabolism , Glutamic Acid/metabolism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Neurons/metabolism , Rats , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Receptors, Presynaptic/drug effects , Receptors, Presynaptic/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
19.
J Comp Neurol ; 515(3): 269-94, 2009 Jul 20.
Article in English | MEDLINE | ID: mdl-19425077

ABSTRACT

In mice and in young adult humans, the subventricular zone (SVZ) contains multipotent, dividing astrocytes, some of which, when cultured, produce neurospheres that differentiate into neurons and glia. It is unknown whether the SVZ of very old humans has this capacity. Here, we report that neural stem/progenitor cells can also be cultured from rapid autopsy samples of SVZ from elderly human subjects, including patients with age-related neurologic disorders. Histological sections of SVZ from these cases showed a glial fibrillary acidic protein (GFAP)-positive ribbon of astrocytes similar to the astrocyte ribbon in human periventricular white matter biopsies that is reported to be a rich source of neural progenitors. Cultures of the SVZ contained 1) neurospheres with a core of Musashi-1-, nestin-, and nucleostemin-immunopositive cells as well as more differentiated GFAP-positive astrocytes; 2) SMI-311-, MAP2a/b-, and beta-tubulin(III)-positive neurons; and 3) galactocerebroside-positive oligodendrocytes. Neurospheres continued to generate differentiated progeny for months after primary culturing, in some cases nearly 2 years postinitial plating. Patch clamp studies of differentiated SVZ cells expressing neuron-specific antigens revealed voltage-dependent, tetrodotoxin-sensitive, inward Na+ currents and voltage-dependent, delayed, slowly inactivating K+ currents, electrophysiologic characteristics of neurons. A subpopulation of these cells also exhibited responses consistent with the kinetics and pharmacology of the h-current. However, although these cells displayed some aspects of neuronal function, they remained immature, insofar as they did not fire action potentials. These studies suggest that human neural progenitor activity may remain viable throughout much of the life span, even in the face of severe neurodegenerative disease.


Subject(s)
Autopsy , Cerebral Ventricles/cytology , Neurodegenerative Diseases , Stem Cells/physiology , Aged , Aged, 80 and over , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cerebral Ventricles/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Middle Aged , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/cytology , Neurons/physiology , Patch-Clamp Techniques , Stem Cells/cytology
20.
J Physiol ; 587(2): 345-61, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19047205

ABSTRACT

Dopaminergic (DAergic) neuronal activity in the ventral tegmental area (VTA) is thought to contribute generally to pleasure, reward, and drug reinforcement and has been implicated in nicotine dependence. nAChRs expressed in the VTA exhibit diverse subunit compositions, but the functional and pharmacological properties are largely unknown. Here, using patch-clamp recordings in single DAergic neurons freshly dissociated from rat VTA, we clarified three functional subtypes of nAChRs (termed ID, IID and IIID receptors) based on whole-cell current kinetics and pharmacology. Kinetic analysis demonstrated that comparing to ID, IID receptor-mediated current had faster activation and decay constant and IIID receptor-mediated current had larger current density. Pharmacologically, ID receptor-mediated current was sensitive to the alpha4beta2-nAChR agonist RJR-2403 and antagonist dihydro-beta-erythroidine (DHbetaE); IID receptor-mediated current was sensitive to the selective alpha7-nAChR agonist choline and antagonist methyllycaconitine (MLA); while IIID receptor-mediated current was sensitive to the beta4-containing nAChR agonist cytisine and antagonist mecamylamine (MEC). The agonist concentration-response relationships demonstrated that IID receptor-mediated current exhibited the highest EC(50) value compared to ID and IIID receptors, suggesting a relatively low agonist affinity of type IID receptors. These results suggest that the type ID, IID and IIID nAChR-mediated currents are predominately mediated by activation of alpha4beta2-nAChR, alpha7-nAChR and a novel nAChR subtype(s), respectively. Collectively, these findings indicate that the VTA DAergic neurons express diversity and multiplicity of functional nAChR subtypes. Interestingly, each DAergic neuron predominantly expresses only one particularly functional nAChR subtype, which may have distinct but important roles in regulation of VTA DA neuronal function, DA transmission and nicotine dependence.


Subject(s)
Dopamine/metabolism , Neurons/physiology , Receptors, Nicotinic/physiology , Ventral Tegmental Area/physiology , Acetylcholine/pharmacology , Aconitine/analogs & derivatives , Aconitine/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Alkaloids/pharmacology , Animals , Azocines/pharmacology , Brain/metabolism , Choline/pharmacology , Dihydro-beta-Erythroidine/pharmacology , Dopamine/pharmacology , Dose-Response Relationship, Drug , Gene Expression/drug effects , Kinetics , Mecamylamine/pharmacology , Neurons/cytology , Neurons/drug effects , Nicotine/analogs & derivatives , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/physiology , Quinolizines/pharmacology , Rats , Rats, Wistar , Receptors, Nicotinic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...