Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Signal Transduct Target Ther ; 7(1): 170, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35641478

ABSTRACT

Cerebellar ataxias are characterized by a progressive decline in motor coordination, but the specific output circuits and underlying pathological mechanism remain poorly understood. Through cell-type-specific manipulations, we discovered a novel GABAergic Purkinje cell (PC) circuit in the cerebellar IV/V lobe that projected to CaMKIIα+ neurons in the fastigial nucleus (FN), which regulated sensorimotor coordination. Furthermore, transcriptomics profiling analysis revealed various cerebellar neuronal identities, and we validated that biorientation defective 1 (BOD1) played an important role in the circuit of IV/V lobe to FN. BOD1 deficit in PCs of IV/V lobe attenuated the excitability and spine density of PCs, accompany with ataxia behaviors. Instead, BOD1 enrichment in PCs of IV/V lobe reversed the hyperexcitability of CaMKIIα+ neurons in the FN and ameliorated ataxia behaviors in L7-Cre; BOD1f/f mice. Together, these findings further suggest that specific regulation of the cerebellar IV/V lobePCs → FNCaMKIIα+ circuit might provide neuromodulatory targets for the treatment of ataxia behaviors.


Subject(s)
Cerebellar Nuclei , Purkinje Cells , Animals , Ataxia , Cerebellar Nuclei/physiology , Mice , Neurons , Purkinje Cells/physiology
2.
Pharmacol Res Perspect ; 10(3): e00947, 2022 06.
Article in English | MEDLINE | ID: mdl-35599337

ABSTRACT

The prolonged exposure to obesogenic diets disrupts the mesocortical dopaminergic input to the prefrontal cortex (PFC). This leads to suboptimal dopamine levels in this brain region, which affects cognition and control of food intake. Treatments that restore mesocortical dopaminergic neurotransmission may improve obesity-associated cognitive dysfunction and modulate food intake to induce weight loss. Given the complexity and multifactorial nature of obesity, combination treatments would likely achieve sizeable and sustained body weight loss and improve obesity-linked outcomes, such as cognitive dysfunction. Given this background, we hypothesize that concomitant activation of serotonin 5-HT2C and histamine H1 receptors, coupled with antagonism of histamine H3 receptors, synergistically modulates mesocortical dopamine neurotransmission and ameliorates obesity-induced cognitive dysfunction. We propose to test the hypothesis in a diet-induced obesity (DIO) rat model by treating animals with the 5-HT2C agonist lorcaserin and the H1 agonist and H3 antagonist betahistine. Consistent with our hypothesis, both lorcaserin and betahistine have been shown to reduce body weight in humans with obesity and animals. Both drugs have been demonstrated to improve cognitive functions by influencing dopaminergic signaling in the PFC. The proposed combination treatment addresses the paucity of studies on obesity treatments that improve cognitive function. This research may also help identify a potential targetable mechanism connecting obesity and neurocognitive outcomes.


Subject(s)
Betahistine , Cognitive Dysfunction , Animals , Benzazepines , Betahistine/pharmacology , Betahistine/therapeutic use , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Dopamine , Obesity/complications , Obesity/drug therapy , Rats , Rats, Sprague-Dawley
3.
Medicine (Baltimore) ; 100(47): e27583, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34964726

ABSTRACT

BACKGROUND: Osteoarthritis (OA) has been identified as a common musculoskeletal condition. As a chronic condition, OA adversely impact the hip and knee joints. Surgical treatment for hip and knee osteoarthritis is associated with high financial and long recovery processes. Therefore, patients are continually searching for alternative methods of treatment. Diacerein is regarded as symptom-modifying, slow-acting drug that could most likely change the disease structure of OA. The present systematic review protocol explains methods utilized to evaluate the clinical therapeutic effects of combining diacerein and glucosamine to treat OA. METHODS: The authors will conduct a search for randomized controlled trials comparing diacerein plus glucosamine with diacerein alone, glucosamine alone, or another treatment in patients with OA. The search will be done in the following online-based databases: EMBASE, MEDLINE, Cochrane Library, Web of Science, China National Knowledge Infrastructure, and WanFang Database. All related RCTs included from inception to September 29, 2021 are included. Two authors will independently conduct data abstraction and quality assessment, and the comparative analysis will compare the results. The present meta-analysis will be performed with the RevMan software (version 5.3), where the results will be expressed as relative risk, mean differences, or standardized mean differences with 95% confidence intervals. RESULTS: This study will be conducted to evaluate the clinical therapeutic effects of combined diacerein and glucosamine in the treatment of OA. CONCLUSION: The summary presented in the study will ascertain whether diacerein plus glucosamine intervention is an efficient and feasible method of treatment for OA patients. TRIAL REGISTRATION NUMBER: 10.17605/OSF.IO/VHPZC.


Subject(s)
Anthraquinones/therapeutic use , Glucosamine/therapeutic use , Osteoarthritis, Hip/drug therapy , Osteoarthritis, Knee/drug therapy , Humans , Meta-Analysis as Topic , Systematic Reviews as Topic
4.
Front Pharmacol ; 12: 728729, 2021.
Article in English | MEDLINE | ID: mdl-34489713

ABSTRACT

Cortical neurons oscillate between Up and Down states during slow wave sleep and general anesthesia. Recent studies show that Up/Down oscillations also occur during quiet wakefulness. Arousal eliminates Down states and transforms Up/Down oscillations to a persistent Up state. Further evidence suggests that Up/Down oscillations are crucial to memory consolidation, whereas their transition to a persistent Up state is essential for arousal and attention. We have shown that D-amphetamine promotes cortical Up state, and the effect depends on activation of central α1A adrenergic receptors. Here, we report that dopamine also plays a role in D-amphetamine's effect. Thus, using local-field-potential recording in the prefrontal cortex in chloral hydrate-anesthetized rats, we showed that the Up-state promoting effect of D-amphetamine was attenuated by antagonists at either D1 or D2-like dopamine receptors. The effect was also partially mimicked by co-activation of D1 and D2-like receptors. These results are consistent with the fact that D-amphetamine increases the release of both norepinephrine and dopamine. They are also in agreement with studies showing that dopamine promotes wakefulness and mediates D-amphetamine-induced emergence from general anesthesia. The effect of D-amphetamine was not mimicked, however, by activation of either D1 or D2-like receptors alone, indicating an interdependence between D1 and D2-like receptors. The dopamine/norepinephrine precursor L-DOPA also failed to promote the Up state. While more studies are needed to understand the difference between L-DOPA and D-amphetamine, our finding may provide an explanation for why L-DOPA lacks significant psychostimulant properties and is ineffective in treating attention-deficit/hyperactivity disorder.

5.
Addict Biol ; 26(1): e12879, 2021 01.
Article in English | MEDLINE | ID: mdl-32003119

ABSTRACT

Cortical neurons oscillate synchronously between the Up and Down state during slow-wave sleep and general anesthesia. Using local-field-potential recording in the rat prefrontal cortex (PFC), we have shown that systemic administration of methylphenidate promotes PFC Up states and reduces PFC slow oscillation, suggesting a depolarizing effect of the drug on PFC neurons. Here, we report that systemic injection of d-amphetamine produced similar effects. Our evidence further suggests that norepinephrine (NE) plays a major role in the effects of d-amphetamine since they were mimicked by the NE reuptake inhibitors tomoxetine and nisoxetine and completely blocked by the α1 receptor antagonist prazosin. The effects of d-amphetamine persisted, however, in the presence of α2 or ß receptor blockade. Experiments with α1 subtype-selective antagonists further suggest that d-amphetamine's effects depend on activation of central, but not peripheral, α1A receptors. Unexpectedly, the putative α1 receptor agonist cirazoline failed to mimic the effects of d-amphetamine. Previous studies suggest that cirazoline is also an antagonist at α2 receptors. Furthermore, it is a partial, not full, agonist at α1B and α1D receptors. Whether or not these properties of cirazoline contribute to its failure to mimic d-amphetamine's effects remains to be determined. Methylphenidate and d-amphetamine are two most common medications for attention-deficit/hyperactivity disorder (ADHD). Both, however, are associated with adverse effects including abuse potential and psychotomimetic effects. Further understanding of their mechanisms of action will help develop safer treatments for ADHD and offer new insights into drug addiction and psychosis.


Subject(s)
Amphetamine/pharmacology , Prefrontal Cortex/drug effects , Receptors, Adrenergic/physiology , Animals , Atomoxetine Hydrochloride/pharmacology , Dextroamphetamine/pharmacology , Dopamine , Male , Methylphenidate/pharmacology , Neurons/drug effects , Norepinephrine/metabolism , Prazosin , Rats
6.
Eur J Pharmacol ; 892: 173826, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33347825

ABSTRACT

Finding alternative treatments for attention-deficit/hyperactivity disorder (ADHD) is crucial given the safety and efficacy problems of current ADHD medications. Droxidopa, also known as L-threo-dihydroxyphenylserine (L-DOPS), is a norepinephrine prodrug that enhances brain norepinephrine and dopamine levels. In this study, we used electrophysiological tests to examine effects of L-DOPS on the prefrontal cortex (PFC) and dopamine neurons in the ventral tegmental area. We also conducted behavioral tests to assess L-DOPS' effects on ADHD-like behaviors in rats. In chloral hydrate-anesthetized rats, PFC local field potentials oscillated between the active, depolarized UP state and the hyperpolarized DOWN state. Mimicking the effect of d-amphetamine, L-DOPS, given after the peripheral amino acid decarboxylase inhibitor, benserazide (BZ), increased the amount of time the PFC spent in the UP state, indicating an excitatory effect of L-DOPS on PFC neurons. Like d-amphetamine, L-DOPS also inhibited dopamine neurons, an effect significantly reversed by the D2-like receptor antagonist raclopride. In the behavioral tests, BZ + L-DOPS improved hyperactivity, inattention and impulsive action of the adolescent spontaneously hypertensive rat (SHR/NCrl), well-validated animal model of the combined type of ADHD. BZ + L-DOPS also reduced impulsive choice and impulsive action of Wistar rats, but did not ameliorate the inattentiveness of Wistar Kyoto rats (WKY/NCrl), proposed model of the ADHD-predominantly inattentive type. In conclusion, L-DOPS produced effects on the PFC and dopamine neurons characteristic of drugs used to treat ADHD. BZ + L-DOPS ameliorated ADHD-like behaviors in rats suggesting its potential as an alternative ADHD treatment.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Behavior, Animal/drug effects , Dopamine Agents/pharmacology , Dopaminergic Neurons/drug effects , Droxidopa/pharmacology , Prefrontal Cortex/drug effects , Ventral Tegmental Area/drug effects , Animals , Attention/drug effects , Attention Deficit Disorder with Hyperactivity/metabolism , Attention Deficit Disorder with Hyperactivity/physiopathology , Attention Deficit Disorder with Hyperactivity/psychology , Benserazide/pharmacology , Delay Discounting/drug effects , Disease Models, Animal , Dopaminergic Neurons/metabolism , Drug Therapy, Combination , Locomotion/drug effects , Male , Maze Learning/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Sprague-Dawley , Species Specificity , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiopathology
7.
Pharmacol Ther ; 212: 107580, 2020 08.
Article in English | MEDLINE | ID: mdl-32454050

ABSTRACT

Hyperkinetic movement disorders comprise a variety of conditions characterized by involuntary movements, which include but are not limited to tardive dyskinesia, chorea associated with Huntington's Disease, and tic disorders. The class of medications that have been used to treat these conditions includes Vesicular Monoamine Transporter-2 (VMAT2) inhibitors. In 2008, the FDA approved tetrabenazine as a treatment for chorea associated with Huntington's Disease. Optimization of the pharmacology of tetrabenazine has since led to the approval of two new VMAT2 inhibitors, deutetrabenazine and valbenazine. The objective of this review is to provide background on the role of VMAT in monoamine neurotransmission, the mechanism of VMAT2 inhibition on the treatment of hyperkinetic disorders (specifically tardive dyskinesia and chorea associated with Huntington's Disease), the pharmacology and pharmacokinetics of the commercially available VMAT2 inhibitors, and a summary of the clinical data to support application of these medications.


Subject(s)
Chorea/drug therapy , Hyperkinesis/drug therapy , Tardive Dyskinesia/drug therapy , Vesicular Monoamine Transport Proteins/antagonists & inhibitors , Animals , Clinical Trials as Topic , Dopamine/metabolism , Humans , Huntington Disease/complications , Synaptic Transmission , Tetrabenazine/analogs & derivatives , Tetrabenazine/therapeutic use , Vesicular Monoamine Transport Proteins/physiology
8.
J Exp Med ; 217(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31699822

ABSTRACT

Blood-brain barrier (BBB) dysfunction has been suggested to play an important role in epilepsy. However, the mechanism mediating the transition from cerebrovascular damage to epilepsy remains unknown. Here, we report that endothelial cyclin-dependent kinase 5 (CDK5) is a central regulator of neuronal excitability. Endothelial-specific Cdk5 knockout led to spontaneous seizures in mice. Knockout mice showed increased endothelial chemokine (C-X-C motif) ligand 1 (Cxcl1) expression, decreased astrocytic glutamate reuptake through the glutamate transporter 1 (GLT1), and increased glutamate synaptic function. Ceftriaxone restored astrocytic GLT1 function and inhibited seizures in endothelial Cdk5-deficient mice, and these effects were also reversed after silencing Cxcl1 in endothelial cells and its receptor chemokine (C-X-C motif) receptor 2 (Cxcr2) in astrocytes, respectively, in the CA1 by AAV transfection. These results reveal a previously unknown link between cerebrovascular factors and epileptogenesis and provide a rationale for targeting endothelial signaling as a potential treatment for epilepsy.


Subject(s)
Chemokine CXCL1/metabolism , Cyclin-Dependent Kinase 5/metabolism , Endothelial Cells/metabolism , Epilepsy/metabolism , Gliosis/metabolism , Receptors, Interleukin-8B/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/metabolism , Cells, Cultured , Endothelial Cells/pathology , Epilepsy/pathology , Gliosis/pathology , Glutamic Acid/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Seizures/metabolism , Seizures/pathology , Signal Transduction/physiology
9.
Theranostics ; 9(20): 5937-5955, 2019.
Article in English | MEDLINE | ID: mdl-31534530

ABSTRACT

Prolonged occlusion of multiple microvessels causes microvascular injury. G protein-coupled receptor 124 (GPR124) has been reported to be required for maintaining central nervous system (CNS) angiogenesis and blood-brain barrier integrity. However, the molecular mechanisms by which GPR124 regulates pericytes during ischemia have remained elusive. Methods: A microsphere embolism-induced ischemia model was used to evaluate the expression of GPR124 following microsphere embolism. Immunocytochemistry and stochastic optical reconstruction microscopy imaging were used to assess the expression and distribution of GPR124 in human brain vascular pericytes (HBVPs) and after the treatment with 3-morpholino-sydnonimine (SIN-1) or oxygen-glucose deprivation (OGD). The effect of GPR124 knockdown or overexpression on HBVP migration was analyzed in vitro using wound healing assays and a microfluidic device. GPR124 loss-of-function studies were performed in HBVPs and HEK293 cells using CRISPR-Cas9-mediated gene deletion. Time-lapse imaging was used to assess dynamic changes in the formation of filopodia in an individual cell. Finally, to explore the functional domains required for GPR124 activity, deletion mutants were constructed for each of the N-terminal domains. Results: GPR124 expression was increased in pericytes following microsphere embolism. Morphological analysis showed localization of GPR124 to focal adhesions where GPR124 bound directly to the actin binding protein vinculin and upregulated Cdc42. SIN-1 or OGD treatment redistributed GPR124 to the leading edges of HBVPs where GPR124 signaling was required for pericyte filopodia formation and directional migration. Partial deletion of GPR124 domains decreased SIN-1-induced filopodia formation and cell migration. Conclusion: Taken together, our results provide the first evidence for a role of GPR124 in pericyte migration under ischemic conditions and suggest that GPR124 was essential for Cdc42 activation and filopodia formation.


Subject(s)
Brain Ischemia/metabolism , Cell Polarity/physiology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Pericytes/cytology , Pericytes/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blotting, Western , Cell Line , Cell Polarity/genetics , Focal Adhesions/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Lentivirus/genetics , Male , Mice , Plasmids/genetics , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Wound Healing/genetics , Wound Healing/physiology
10.
Mol Psychiatry ; 24(10): 1461-1477, 2019 10.
Article in English | MEDLINE | ID: mdl-30886335

ABSTRACT

Anxiety disorders are the most prevalent psychiatric disorders, but their pathogenic mechanism remains poorly understood. Here, we report that transmembrane protein 74 (TMEM74), which contains two putative transmembrane domains and exhibits high levels of mRNA in the brain, is closely associated with the pathogenesis of anxiety disorders. TMEM74 was decreased in the serum of patients with anxiety and the basolateral amygdaloid nucleus (BLA) in chronic stress mice. Furthermore, genetic deletion of Tmem74 or selective knockdown of Tmem74 in BLA pyramidal neurons resulted in anxiety-like behaviors in mice. Whole-cell recordings in BLA pyramidal neurons revealed lower hyperpolarization-activated cation current (Ih) and greater input resistance and excitability in Tmem74-/- neurons than in wild-type neurons. Accordingly, surface expression of hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels was also lower in the BLA of Tmem74-/- mice. The Ih current blocker ZD7288 mimicked these effects in BLA pyramidal neurons in wild-type mice but not in Tmem74-/- mice. Consistent with the improvement in anxiety-like behaviors, Tmem74 overexpression restored HCN1 channel trafficking and pyramidal neuron excitability in the BLA of Tmem74-/- and chronic stress mice. Mechanistically, we demonstrate that interactions between Tmem74 and HCN1 are physiologically relevant and that transmembrane domain 1 (TM1) is essential for the cellular membrane localization of Tmem74 to enhance Ih. Together, our findings suggest that Tmem74 coupling with HCN1 acts as a critical component in the pathophysiology of anxiety and is a potential target for new treatments of anxiety disorders.


Subject(s)
Anxiety/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Membrane Proteins/metabolism , Animals , Anxiety/genetics , Anxiety Disorders/genetics , Anxiety Disorders/metabolism , Basolateral Nuclear Complex/metabolism , Brain/metabolism , Cyclic Nucleotide-Gated Cation Channels , Hippocampus/metabolism , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Membrane Potentials/physiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Patch-Clamp Techniques , Potassium Channels/genetics , Protein Transport , Pyramidal Cells/metabolism
11.
Neuron ; 101(5): 920-937.e13, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30685224

ABSTRACT

The proper interactions between blood vessels and neurons are critical for maintaining the strength of neural circuits and cognitive function. However, the precise molecular events underlying these interactions remain largely unknown. Here, we report that the selective knockout of semaphorin 3G (Sema3G) in endothelial cells impaired hippocampal-dependent memory and reduced dendritic spine density in CA1 neurons in mice; these effects were reversed after restoration of Sema3G levels in the hippocampus by AAV transfection. We further show that Sema3G increased excitatory synapse density via neuropilin-2/PlexinA4 signaling and through activation of Rac1. These results provide the first evidence that, in the central nervous system, endothelial Sema3G serves as a vascular-derived synaptic organizer that regulates synaptic plasticity and hippocampal-dependent memory. Our findings highlight the role of vascular endothelial cells in regulating cognitive function through intercellular communication with neurons in the hippocampus.


Subject(s)
Endothelium, Vascular/metabolism , Hippocampus/metabolism , Memory Disorders/metabolism , Neuronal Plasticity , Semaphorins/metabolism , Animals , Cells, Cultured , Female , HEK293 Cells , Hippocampus/physiology , Humans , Male , Memory Disorders/genetics , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-2/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Semaphorins/genetics , Synapses/metabolism , Synapses/physiology
12.
Neuroscience ; 384: 178-187, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29859979

ABSTRACT

Indirect evidence suggests that low doses of ketamine disinhibit (excite) pyramidal neurons in the prefrontal cortex (PFC). In this study, we directly examined the effect of ketamine on PFC pyramidal neurons using simultaneous single-cell and local-field-potential (LFP) recording in chloral hydrate-anesthetized rats. In all animals studied, PFC LFPs showed oscillations (0.3-1.5 Hz) between the active UP state and the relatively quiescent DOWN state, and pyramidal neurons fired preferentially during the UP state. Ketamine (1.25-20 mg/kg, i.v.) inhibited 80% of cells tested and consistently shifted PFC LFPs toward the DOWN state. The inhibitory effect of ketamine was mimicked by MK801, but not by the NR2B-selective NMDA receptor antagonist Ro25-6981. It was not blocked by the dopamine receptor antagonist fluphenazine, the GABAA receptor antagonist picrotoxinin, or the GABAB receptor antagonist CGP46381. These results are consistent with the high density of NMDA receptors expressed on PFC pyramidal neurons and our previous studies showing that blockade of NMDA receptors by ketamine inhibits dendritic NMDA receptor-mediated bursting in PFC pyramidal neurons. Thus, in addition to the previously proposed disinhibitory effect mediated through PFC interneurons, our data suggest that ketamine has an inhibitory effect on PFC pyramidal neurons. Our evidence further suggests that the effect is mediated through non-NR2B-containing NMDA receptors, independent of ketamine's effect on dopamine and GABA transmission. Further understanding of the two opposing effects of ketamine on PFC pyramidal neurons may provide important new insights into its mechanism of action.


Subject(s)
Action Potentials/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Animals , Dizocilpine Maleate/pharmacology , Dopamine Antagonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , GABA-B Receptor Antagonists/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
13.
Sci Rep ; 8(1): 3274, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29459754

ABSTRACT

The initiation of psychostimulant sensitization depends on the mesocorticolimbic dopamine (DA) system. Although many cellular adaptations has been reported to be associated with this addictive behavior, the overall influence of these adaptations on the network regulation of DA neurons has not been established. Here, we profile a network-driven slow oscillation (SO) in the firing activity of ventral tegmental area (VTA) putative DA and non-DA neurons and their correlation with locomotor sensitization induced by repeated administration of cocaine. One day after the last cocaine injection, the power of SO (Pso) significantly increased both in DA and non-DA neurons. Interestingly, the Pso in DA neurons was positively correlated, while Pso in non-DA neurons was negatively correlated with the level of locomotor sensitization. On the other hand, the firing rates of DA and non-DA neurons were both elevated, but none exhibited any correlation with the level of sensitization. Fourteen days after the last injection, the Pso of DA neurons dissipated but still positively correlated with the level of sensitization. In contrast, the Pso in non-DA neurons lost correlation with locomotor sensitization. These results suggest that cocaine-induced locomotor sensitization is associated with long-term network adaptation in DA system and that DA and non-DA neurons may corporately facilitate/hamper the initiation of locomotor sensitization.


Subject(s)
Action Potentials , Cocaine/metabolism , Dopamine Uptake Inhibitors/metabolism , Neurons/drug effects , Ventral Tegmental Area/drug effects , Animals , Behavior, Animal , Male , Motor Activity , Rats, Sprague-Dawley
14.
Neuropharmacology ; 131: 431-439, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29339293

ABSTRACT

Amphetamine-like psychostimulants, including methylphenidate, have been shown to produce two opposing effects on dopamine (DA) neurons: a DA receptor-mediated feedback inhibition and a non-DA receptor-mediated excitation. To test whether the latter effect is mediated through the prefrontal cortex (PFC), we made dual-site recordings from the PFC and ventral tegmental area (VTA). Consistent with previous reports, methylphenidate inhibited VTA DA neurons. The D2 receptor antagonist raclopride completely reversed the inhibition and further increased the activity, particularly bursting, to above pre-drug baseline. This increase in DA cell activity was blocked by the α1 receptor antagonist prazosin, suggesting an effect mediated through α1 receptors. Recordings in the PFC showed that methylphenidate increased PFC UP state duration and shifted the functional coupling between the PFC and DA neurons from negative to positive. The former effect was partially reversed by not only prazosin, but also raclopride, whereas the latter was reversed only by raclopride. These results suggest that methylphenidate increases PFC cell activity through both α1 and D2 receptors. Its effect on PFC-DA cell functional coupling, however, is mediated through D2 receptors. The finding that the latter effect was unaffected by prazosin further suggests that it does not play a significant role in the α1-mediated excitatory effect of methylphenidate on DA neurons. However, the shift in PFC-DA cell functional coupling from negative to positive may significantly alter the relative timing between DA and glutamate release from DA and PFC terminals and thus the synaptic plasticity that depends on DA-glutamate interaction.


Subject(s)
Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/drug effects , Methylphenidate/pharmacology , Prefrontal Cortex/drug effects , Ventral Tegmental Area/drug effects , Animals , Dopaminergic Neurons/physiology , Dose-Response Relationship, Drug , Male , Neural Pathways/drug effects , Neural Pathways/physiology , Prefrontal Cortex/physiology , Rats, Sprague-Dawley , Receptors, Dopamine D2/metabolism , Ventral Tegmental Area/physiology
15.
Med Hypotheses ; 93: 113-6, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27372868

ABSTRACT

Clinical evidence suggests that the prefrontal cortex (PFC) is hypofunctional in disorders including schizophrenia, drug addiction, and attention-deficit/hyperactivity disorder (ADHD). In schizophrenia, hypofrontality has been further suggested to cause both the negative and cognitive symptoms, and overactivity of dopamine neurons that project to subcortical areas. The latter may contribute to the development of positive symptoms of the disorder. Nevertheless, what causes hypofrontality and how it alters dopamine transmission in subcortical structures remain unclear due, in part, to the difficulty in modeling hypofrontality using previous techniques (e.g. PFC lesioning, focal cooling, repeated treatment with psychotomimetic drugs). We propose that the use of designer receptors exclusively activated by designer drugs (DREADDs) chemogenetic technique will allow precise interrogations of PFC functions. Combined with electrophysiological recordings, we can investigate the effects of PFC hypofunction on activity of dopamine neurons. Importantly, from a drug target discovery perspective, the use of DREADDs will enable us to examine whether chemogenetically enhancing PFC activity will reverse the behavioral abnormalities associated with PFC hypofunction and dopamine neuron overactivity, and also explore druggable targets for the treatment of schizophrenia and other disorders associated with abnormalities via modulation of the G-protein coupled receptor signaling pathway. In conclusion, the use of the DREADDs technique has several advantages over other previously employed strategies to simulate PFC hypofunction not only in terms of disease modeling but also from the viewpoint of drug target discovery.


Subject(s)
Prefrontal Cortex/physiopathology , Animals , Attention Deficit Disorder with Hyperactivity/physiopathology , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Drug Delivery Systems , Electrophysiology , Humans , Mice , Models, Theoretical , Rats , Schizophrenia/physiopathology , Signal Transduction , Substance-Related Disorders/physiopathology
16.
Eur J Pharmacol ; 764: 562-570, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26209364

ABSTRACT

The precise mechanisms by which cocaine and amphetamine-like psychostimulants exert their reinforcing effects are not yet fully defined. It is widely believed, however, that these drugs produce their effects by enhancing dopamine neurotransmission in the brain, especially in limbic areas such as the nucleus accumbens, by inducing dopamine transporter-mediated reverse transport and/or blocking dopamine reuptake though the dopamine transporter. Here, we present the evidence that aside from dopamine transporter, non-dopamine transporter-mediated mechanisms also participate in psychostimulant-induced dopamine release and contribute to the behavioral effects of these drugs, such as locomotor activation and reward. Accordingly, psychostimulants could increase norepinephrine release in the prefrontal cortex, the latter then alters the firing pattern of dopamine neurons resulting in changes in action potential-dependent dopamine release. These alterations would further affect the temporal pattern of dopamine release in the nucleus accumbens, thereby modifying information processing in that area. Hence, a synaptic input to a nucleus accumbens neuron may be enhanced or inhibited by dopamine depending on its temporal relationship to dopamine release. Specific temporal patterns of dopamine release may also be required for certain forms of synaptic plasticity in the nucleus accumbens. Together, these effects induced by psychostimulants, mediated through a non-dopamine transporter-mediated mechanism involving norepinephrine and the prefrontal cortex, may also contribute importantly to the reinforcing properties of these drugs.


Subject(s)
Amphetamine/pharmacology , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Cocaine/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Action Potentials , Adrenergic Neurons/drug effects , Adrenergic Neurons/metabolism , Amphetamine/metabolism , Animals , Brain/metabolism , Central Nervous System Stimulants/metabolism , Cocaine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/metabolism , Humans , Kinetics , Neuronal Plasticity , Norepinephrine/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
17.
Cell Res ; 25(6): 674-90, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25998681

ABSTRACT

Septic encephalopathy (SE) is a critical factor determining sepsis mortality. Vascular inflammation is known to be involved in SE, but the molecular events that lead to the development of encephalopathy remain unclear. Using time-lapse in vivo two-photon laser scanning microscopy, we provide the first direct evidence that cecal ligation and puncture in septic mice induces microglial trafficking to sites adjacent to leukocyte adhesion on inflamed cerebral microvessels. Our data further demonstrate that septic injury increased the chemokine CXCL1 level in brain endothelial cells by activating endothelial P2RX7 and eventually enhanced the binding of Mac-1 (CD11b/CD18)-expressing leukocytes to endothelial ICAM-1. In turn, leukocyte adhesion upregulated endothelial CX3CL1, thereby triggering microglia trafficking to the injured site. The sepsis-induced increase in endothelial CX3CL1 was abolished in CD18 hypomorphic mutant mice. Inhibition of the P2RX7 pathway not only decreased endothelial ICAM-1 expression and leukocyte adhesion but also prevented microglia overactivation, reduced brain injury, and consequently doubled the early survival of septic mice. These results demonstrate the role of the P2RX7 pathway in linking neurovascular inflammation to brain damage in vivo and provide a rationale for targeting endothelial P2RX7 for neurovascular protection during SE.


Subject(s)
Brain Injuries/metabolism , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/metabolism , Macrophage-1 Antigen/metabolism , Receptors, Purinergic P2X7/metabolism , Sepsis-Associated Encephalopathy/metabolism , Animals , Brain Injuries/pathology , Cell Adhesion , Endothelial Cells/pathology , Leukocytes/pathology , Mice , Mice, Mutant Strains , Sepsis-Associated Encephalopathy/pathology
18.
Synapse ; 69(1): 41-51, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25196543

ABSTRACT

The rat globus pallidus (GP) is homologous to the primate GP externus. Studies with injectable anesthetics suggest that GP neurons can be classified into Type-I and Type-II cells based on extracellularly recorded spike shape, or positively coupled (PC), negatively coupled (NC), and uncoupled (UC) cells based on functional connectivity with the cortex. In this study, we examined the electrophysiology of rat GP neurons using the inhalational anesthetic isoflurane which offers more constant and easily regulated levels of anesthesia than injectable anesthetics. In 130 GP neurons recorded using small-tip glass electrodes (<1 µm), all but one fired Type-II spikes (positive/negative waveform). Type-I cells were unlikely to be inhibited by isoflurane since all GP neurons also fired Type-II spikes under ketamine-induced anesthesia. When recorded with large-tip electrodes (∼2 µm), however, over 70% of GP neurons exhibited Type-I spikes (negative/positive waveform). These results suggest that the spike shape, recorded extracellularly, varies depending on the electrode used and is not reliable in distinguishing Type-I and Type-II neurons. Using dual-site recording, 40% of GP neurons were identified as PC cells, 17.5% NC cells, and 42.5% UC cells. The three subtypes also differed significantly in firing rate and pattern. Lesions of dopamine neurons increased the number of NC cells, decreased that of UC cells, and significantly shifted the phase relationship between PC cells and the cortex. These results support the presence of GP neuron subtypes and suggest that each subtype plays a different role in the pathophysiology of Parkinson's disease. Synapse 69:41-51, 2015. © 2014 Wiley Periodicals, Inc.


Subject(s)
Dopamine/deficiency , Globus Pallidus/physiology , Neurons/classification , Neurons/physiology , Action Potentials/drug effects , Action Potentials/physiology , Anesthetics, Inhalation/pharmacology , Animals , Dopamine/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Globus Pallidus/drug effects , Globus Pallidus/physiopathology , Isoflurane/pharmacology , Ketamine/pharmacology , Male , Microelectrodes , Motor Cortex/drug effects , Motor Cortex/physiology , Neural Pathways/drug effects , Neural Pathways/physiology , Neural Pathways/physiopathology , Neurons/drug effects , Oxidopamine , Rats, Sprague-Dawley
19.
J Alzheimers Dis ; 38(1): 93-101, 2014.
Article in English | MEDLINE | ID: mdl-24077431

ABSTRACT

BACKGROUND: Diffusion tensor imaging (DTI) suggests the presence of white matter abnormality at the prodromal stage in human Alzheimer's disease (AD). OBJECTIVE: To use a mouse model of AD to determine whether the white matter abnormality detected by in vivo DTI is associated with functional deficits and axon damage. METHODS: Amyloid-ß1-42 (Aß1-42) was injected into the left lateral ventricle in mice. Two months after the injection, in vivo DTI and visual evoked potential (VEP) recordings were performed, followed by immunohistochemistry of phosphorylated neurofilament and myelin basic protein. RESULTS: DTI of Aß1-42-treated mice showed a significant increase of radial diffusivity in white matter including the optic nerves and tracts. The abnormality was associated with decreased amplitude and increased latency of VEP. Immunohistochemistry confirmed a significant loss of axons and myelin integrity. CONCLUSION: White matter damage induced by Aß1-42 in mice can be detected non-invasively by DTI.


Subject(s)
Amyloid beta-Peptides/toxicity , Diffusion Magnetic Resonance Imaging/methods , Leukoencephalopathies/chemically induced , Leukoencephalopathies/diagnosis , Peptide Fragments/toxicity , Animals , Body Weight/drug effects , Disease Models, Animal , Evoked Potentials, Visual/drug effects , Female , Functional Laterality , Leukoencephalopathies/physiopathology , Male , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Neurofilament Proteins/metabolism , Optic Nerve/pathology
20.
Biochem Pharmacol ; 86(8): 1173-80, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23933294

ABSTRACT

Tobacco use is a major public health problem. Nicotine acts on widely distributed nicotinic acetylcholine receptors (nAChRs) in the brain and excites dopamine (DA) neurons in the ventral tegmental area (VTA). The elicited increase of DA neuronal activity is thought to be an important mechanism for nicotine reward and subsequently the transition to addiction. However, the current understanding of nicotine reward is based predominantly on the data accumulated from in vitro studies, often from VTA slices. Isolated VTA slices artificially terminate communications between neurons in the VTA and other brain regions that may significantly alter nicotinic effects. Consequently, the mechanisms of nicotinic excitation of VTA DA neurons under in vivo conditions have received only limited attention. Building upon the existing knowledge acquired in vitro, it is now time to elucidate the integrated mechanisms of nicotinic reward on intact systems that are more relevant to understanding the action of nicotine or other addictive drugs. In this review, we summarize recent studies that demonstrate the impact of prefrontal cortex (PFC) on the modulation of VTA DA neuronal function and nicotine reward. Based on existing evidence, we propose a new hypothesis that PFC-VTA functional coupling serves as an integration mechanism for nicotine reward. Moreover, addiction may develop due to nicotine perturbing the PFC-VTA coupling and thereby eliminating the PFC-dependent cognitive control over behavior.


Subject(s)
Nicotine/pharmacology , Prefrontal Cortex/physiology , Receptors, Nicotinic/metabolism , Ventral Tegmental Area/physiology , Gene Expression Regulation , Humans , Receptors, Nicotinic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...