Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 4566, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32165725

ABSTRACT

Peripheral biomarker and post-mortem brains studies have shown alterations of neuronal calcium sensor 1 (Ncs-1) expression in people with bipolar disorder or schizophrenia. However, its engagement by psychiatric medications and potential contribution to behavioral regulation remains elusive. We investigated the effect on Ncs-1 expression of valproic acid (VPA), a mood stabilizer used for the management of bipolar disorder. Treatment with VPA induced Ncs-1 gene expression in cell line while chronic administration of this drug to mice increased both Ncs-1 protein and mRNA levels in the mouse frontal cortex. Inhibition of histone deacetylases (HDACs), a known biochemical effect of VPA, did not alter the expression of Ncs-1. In contrast, pharmacological inhibition or genetic downregulation of glycogen synthase kinase 3ß (Gsk3ß) increased Ncs-1 expression, whereas overexpression of a constitutively active Gsk3ß had the opposite effect. Moreover, adeno-associated virus-mediated Ncs-1 overexpression in mouse frontal cortex caused responses similar to those elicited by VPA or lithium in tests evaluating social and mood-related behaviors. These findings indicate that VPA increases frontal cortex Ncs-1 gene expression as a result of Gsk3 inhibition. Furthermore, behavioral changes induced by Ncs-1 overexpression support a contribution of this mechanism in the regulation of behavior by VPA and potentially other psychoactive medications inhibiting Gsk3 activity.


Subject(s)
Anxiety/chemically induced , Frontal Lobe/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Neuronal Calcium-Sensor Proteins/genetics , Neuronal Calcium-Sensor Proteins/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Valproic Acid/adverse effects , Animals , Anxiety/genetics , Anxiety/metabolism , Cell Line , Disease Models, Animal , Down-Regulation , Glycogen Synthase Kinase 3 beta/genetics , HEK293 Cells , Humans , Male , Mice , PC12 Cells , Rats , Social Behavior , Up-Regulation , Valproic Acid/administration & dosage
2.
Nat Commun ; 10(1): 4906, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31659165

ABSTRACT

The delivery of biologic cargoes to airway epithelial cells is challenging due to the formidable barriers imposed by its specialized and differentiated cells. Among cargoes, recombinant proteins offer therapeutic promise but the lack of effective delivery methods limits their development. Here, we achieve protein and SpCas9 or AsCas12a ribonucleoprotein (RNP) delivery to cultured human well-differentiated airway epithelial cells and mouse lungs with engineered amphiphilic peptides. These shuttle peptides, non-covalently combined with GFP protein or CRISPR-associated nuclease (Cas) RNP, allow rapid entry into cultured human ciliated and non-ciliated epithelial cells and mouse airway epithelia. Instillation of shuttle peptides combined with SpCas9 or AsCas12a RNP achieves editing of loxP sites in airway epithelia of ROSAmT/mG mice. We observe no evidence of short-term toxicity with a widespread distribution restricted to the respiratory tract. This peptide-based technology advances potential therapeutic avenues for protein and Cas RNP delivery to refractory airway epithelial cells.


Subject(s)
Bacterial Proteins/metabolism , Drug Delivery Systems/methods , Endonucleases/metabolism , Epithelial Cells/metabolism , Lung Diseases/therapy , Lung/metabolism , Peptides/genetics , Animals , Bacterial Proteins/genetics , Bronchi/cytology , Bronchi/metabolism , Endonucleases/genetics , Genetic Therapy , Humans , Lung Diseases/genetics , Lung Diseases/metabolism , Mice , Peptides/administration & dosage , Peptides/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Swine
3.
PLoS One ; 13(4): e0195558, 2018.
Article in English | MEDLINE | ID: mdl-29617431

ABSTRACT

Delivery of recombinant proteins to therapeutic cells is limited by a lack of efficient methods. This hinders the use of transcription factors or Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) ribonucleoproteins to develop cell therapies. Here, we report a soluble peptide designed for the direct delivery of proteins to mammalian cells including human stem cells, hard-to-modify primary natural killer (NK) cells, and cancer cell models. This peptide is composed of a 6x histidine-rich domain fused to the endosomolytic peptide CM18 and the cell penetrating peptide PTD4. A less than two-minute co-incubation of 6His-CM18-PTD4 peptide with spCas9 and/or asCpf1 CRISPR ribonucleoproteins achieves robust gene editing. The same procedure, co-incubating with the transcription factor HoxB4, achieves transcriptional regulation. The broad applicability and flexibility of this DNA- and chemical-free method across different cell types, particularly hard-to-transfect cells, opens the way for a direct use of proteins for biomedical research and cell therapy manufacturing.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Transcription Factors/administration & dosage , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cricetulus , Cytosol/metabolism , Endocytosis , Escherichia coli , Humans , Mice , Myoblasts/cytology , Myoblasts/metabolism , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism
4.
J Recept Signal Transduct Res ; 35(3): 224-32, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26459714

ABSTRACT

Mood stabilizers are a heterogeneous class of drugs having antidepressant and anti-manic effects in bipolar disorders, depression and schizophrenia. Despite wide clinical applications, the mechanisms underlying their shared actions and therapeutic specificity are unknown. Here, we examine the effects of the structurally unrelated mood stabilizers lamotrigine, lithium and valproate on G protein and beta-arrestin-dependent components of dopamine D2 receptor signaling and assess their contribution to the behavioral effects of these drugs. When administered chronically to mice lacking either D2 receptors or beta-arrestin 2, lamotrigine, lithium and valproate failed to affect Akt/GSK3 signaling as they do in normal littermates. This lack of effect on signaling resulted in a loss of responsiveness to mood stabilizers in tests assessing "antimanic" or "antidepressant"-like behavioral drug effects. This shows that mood stabilizers lamotrigine, lithium and valproate can exert behavioral effects in mice by disrupting the beta-arrestin 2-mediated regulation of Akt/GSK3 signaling by D2 dopamine receptors, thereby suggesting a shared mechanism for mood stabilizer selectivity.


Subject(s)
Arrestins/metabolism , Behavior, Animal/drug effects , Brain/metabolism , Psychotropic Drugs/administration & dosage , Receptors, Dopamine D2/metabolism , Signal Transduction/drug effects , Animals , Brain/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mood Disorders/drug therapy , Mood Disorders/metabolism
5.
Proc Natl Acad Sci U S A ; 112(33): E4610-9, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26240334

ABSTRACT

Inhibition of glycogen synthase kinase 3ß (GSK3ß) is a shared action believed to be involved in the regulation of behavior by psychoactive drugs such as antipsychotics and mood stabilizers. However, little is known about the identity of the substrates through which GSK3ß affects behavior. We identified fragile X mental retardation-related protein 1 (FXR1P), a RNA binding protein associated to genetic risk for schizophrenia, as a substrate for GSK3ß. Phosphorylation of FXR1P by GSK3ß is facilitated by prior phosphorylation by ERK2 and leads to its down-regulation. In contrast, behaviorally effective chronic mood stabilizer treatments in mice inhibit GSK3ß and increase FXR1P levels. In line with this, overexpression of FXR1P in the mouse prefrontal cortex also leads to comparable mood-related responses. Furthermore, functional genetic polymorphisms affecting either FXR1P or GSK3ß gene expression interact to regulate emotional brain responsiveness and stability in humans. These observations uncovered a GSK3ß/FXR1P signaling pathway that contributes to regulating mood and emotion processing. Regulation of FXR1P by GSK3ß also provides a mechanistic framework that may explain how inhibition of GSK3ß can contribute to the regulation of mood by psychoactive drugs in mental illnesses such as bipolar disorder. Moreover, this pathway could potentially be implicated in other biological functions, such as inflammation and cell proliferation, in which FXR1P and GSK3 are known to play a role.


Subject(s)
Affect , Emotions , Gene Expression Regulation , Glycogen Synthase Kinase 3/metabolism , RNA-Binding Proteins/physiology , Adult , Animals , Behavior, Animal , Facial Expression , Female , Genotype , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Movement , Phosphorylation , Polymorphism, Single Nucleotide , Prefrontal Cortex/physiology , Valproic Acid/administration & dosage , Young Adult
6.
Psychopharmacology (Berl) ; 231(1): 109-22, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23912772

ABSTRACT

RATIONALE: Psychostimulants such as amphetamine and methylphenidate, which target the dopamine transporter (DAT), are the most frequently used drugs for the treatment of hyperactivity and cognitive deficits in humans with attention deficit hyperactivity disorder (ADHD). While psychostimulants can increase activity in healthy subjects, they exert a "paradoxical" calming effect in humans with ADHD as well as in hyperactive mice lacking the dopamine transporter (DAT-KO mice). However, the mechanism of action of these drugs and their impact on cognition in the absence of DAT remain poorly understood. OBJECTIVES: This study was conducted to investigate the effects of psychostimulants and noradrenergic and serotonergic drugs on cognition in DAT-KO mice and normal (WT) littermates. METHODS: We used a recently developed behavioral apparatus, the automated H-maze. The H-maze involves the consecutive learning of three different rules: delayed alternation, nonalternation, and reversal tasks. RESULTS: Treatment of WT animals with the psychostimulants replicated the behavior observed in untreated DAT-KO mice while "paradoxically" restoring cognitive performances in DAT-KO mice. Further investigation of the potential involvement of other monoamine systems in the regulation of cognitive functions showed that the norepinephrine transporter blocker atomoxetine restored cognitive performances in DAT-KO mice without affecting hyperactivity. In contrast, the nonselective serotonin receptor agonist 5CT, which antagonizes hyperactivity in DAT-KO mice, had no effect on cognitive functions. CONCLUSIONS: Taken together, these data allow dissociation of the locomotor and cognitive effects of ADHD drugs and suggest that the combination of DAT-KO mice with the automated H-maze can constitute a powerful experimental paradigm for the preclinical development of therapeutic approaches for ADHD.


Subject(s)
Central Nervous System Stimulants/pharmacology , Cognition/drug effects , Dopamine Plasma Membrane Transport Proteins/genetics , Hyperkinesis/psychology , Motor Activity/drug effects , Propylamines/pharmacology , Amphetamine/pharmacology , Analysis of Variance , Animals , Atomoxetine Hydrochloride , Dose-Response Relationship, Drug , Hyperkinesis/genetics , Maze Learning/drug effects , Methamphetamine/pharmacology , Mice , Mice, Inbred C57BL , Norepinephrine/pharmacology , Psychomotor Performance/drug effects , Serotonin/pharmacology , Serotonin Agents/pharmacology , Smell/drug effects
7.
Neuropsychopharmacology ; 39(5): 1125-34, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24196946

ABSTRACT

Polymorphisms in the gene encoding the serotonin synthesis enzyme Tph2 have been identified in mental illnesses, including bipolar disorder, major depression, autism, schizophrenia, and ADHD. Deficits in cognitive flexibility and perseverative behaviors are shared common symptoms in these disorders. However, little is known about the impact of Tph2 gene variants on cognition. Mice expressing a human TPH2 variant (Tph2-KI) were used to investigate cognitive consequences of TPH2 loss of function and pharmacological treatments. We applied a recently developed behavioral assay, the automated H-maze, to study cognitive functions in Tph2-KI mice. This assay involves the consecutive discovery of three different rules: a delayed alternation task, a non-alternation task, and a delayed reversal task. Possible contribution of locomotion, reward, and sensory perception were also investigated. The expression of loss-of-function mutant Tph2 in mice was associated with impairments in reversal learning and cognitive flexibility, accompanied by perseverative behaviors similar to those observed in human clinical studies. Pharmacological restoration of 5-HT synthesis with 5-hydroxytryptophan or treatment with the 5-HT(2C) receptor agonist CP809.101 reduced cognitive deficits in Tph2-KI mice and abolished perseveration. In contrast, treatment with the psychostimulant methylphenidate exacerbated cognitive deficits in mutant mice. Results from this study suggest a contribution of TPH2 in the regulation of cognition. Furthermore, identification of a role for a 5-HT(2) receptor agonist as a cognition-enhancing agent in mutant mice suggests a potential avenue to explore for the personalized treatment of cognitive symptoms in humans with reduced 5-HT synthesis and TPH2 polymorphisms.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Piperazines/pharmacology , Pyrazines/pharmacology , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT2 Receptor Agonists/pharmacology , Tryptophan Hydroxylase/metabolism , 5-Hydroxytryptophan/pharmacology , Animals , Avoidance Learning/physiology , Cognition/drug effects , Cognition/physiology , Cognition Disorders/genetics , Dopamine Uptake Inhibitors/pharmacology , Humans , Maze Learning/drug effects , Maze Learning/physiology , Methylphenidate/pharmacology , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Mutation , Olfactory Perception/physiology , Reversal Learning/drug effects , Reversal Learning/physiology , Reward , Serotonin Agents/pharmacology , Tryptophan Hydroxylase/genetics
8.
Nat Neurosci ; 16(2): 183-92, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23292683

ABSTRACT

A major unresolved issue in treating pain is the paradoxical hyperalgesia produced by the gold-standard analgesic morphine and other opiates. We found that hyperalgesia-inducing treatment with morphine resulted in downregulation of the K(+)-Cl(-) co-transporter KCC2, impairing Cl(-) homeostasis in rat spinal lamina l neurons. Restoring the anion equilibrium potential reversed the morphine-induced hyperalgesia without affecting tolerance. The hyperalgesia was also reversed by ablating spinal microglia. Morphine hyperalgesia, but not tolerance, required µ opioid receptor-dependent expression of P2X4 receptors (P2X4Rs) in microglia and µ-independent gating of the release of brain-derived neurotrophic factor (BDNF) by P2X4Rs. Blocking BDNF-TrkB signaling preserved Cl(-) homeostasis and reversed the hyperalgesia. Gene-targeted mice in which Bdnf was deleted from microglia did not develop hyperalgesia to morphine. However, neither morphine antinociception nor tolerance was affected in these mice. Our findings dissociate morphine-induced hyperalgesia from tolerance and suggest the microglia-to-neuron P2X4-BDNF-KCC2 pathway as a therapeutic target for preventing hyperalgesia without affecting morphine analgesia.


Subject(s)
Chlorides/metabolism , Homeostasis/drug effects , Hyperalgesia/drug therapy , Microglia/drug effects , Morphine/administration & dosage , Narcotics/administration & dosage , Neurons/drug effects , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Hot Temperature/adverse effects , Ion Channel Gating/drug effects , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/physiology , Motor Activity/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pain Threshold/drug effects , Patch-Clamp Techniques , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X4/genetics , Receptors, Purinergic P2X4/metabolism , Ribosome Inactivating Proteins, Type 1/pharmacology , Rotarod Performance Test , Saporins , Signal Transduction/drug effects , Signal Transduction/genetics , Spinal Cord/cytology , Symporters/metabolism , Time Factors , Touch , Vocalization, Animal/drug effects , K Cl- Cotransporters
9.
Front Mol Neurosci ; 4: 38, 2011.
Article in English | MEDLINE | ID: mdl-22065948

ABSTRACT

Brain dopamine receptors have been preferred targets for numerous pharmacological compounds developed for the treatment of various neuropsychiatric disorders. Recent discovery that D2 dopamine receptors, in addition to cAMP pathways, can engage also in Akt/GSK3 signaling cascade provided a new framework to understand intracellular signaling mechanisms involved in dopamine-related behaviors and pathologies. Here we review a recent progress in understanding the role of Akt, GSK3, and related signaling molecules in dopamine receptor signaling and functions. Particularly, we focus on the molecular mechanisms involved, interacting partners, role of these signaling events in the action of antipsychotics, psychostimulants, and antidepressants as well as involvement in pathophysiology of schizophrenia, bipolar disorder, and Parkinson's disease. Further understanding of the role of Akt/GSK3 signaling in dopamine receptor functions could provide novel targets for pharmacological interventions in dopamine-related disorders.

10.
Front Neuroanat ; 5: 58, 2011.
Article in English | MEDLINE | ID: mdl-21922001

ABSTRACT

Multifunctional scaffolding protein beta-arrestins (ßArr) and the G protein-receptor kinases are involved in the desensitization of several G protein-coupled receptors (GPCR). However, arrestins can also contribute to GPCR signaling independently from G proteins. In this review, we focus on the role of ßArr in the regulation of dopamine receptor functions in the striatum. First, we present in vivo evidence supporting a role for these proteins in the regulation of dopamine receptor desensitization. Second, we provide an overview of the roles of ßArr2 in the regulation of extracellular-signal-regulated kinases/MAP kinases and Akt/GSK3 signaling pathways downstream of the D1 and D2 dopamine receptors. Thereafter, we examine the possible involvement of ßArr-mediated signaling in the action of dopaminergic drugs used for the treatment of mental disorders. Finally, we focus on different potential cellular proteins regulated by ßArr-mediated signaling which could contribute to the regulation of behavioral responses to dopamine. Overall, the identification of a cell signaling function for ßArr downstream of dopamine receptors underscores the intricate complexity of the intertwined mechanisms regulating and mediating cell signaling in the basal ganglia. Understanding these mechanisms may lead to a better comprehension of the several roles played by these structures in the regulation of mood and to the development of new psychoactive drugs having better therapeutic efficacy.

11.
Med Sci (Paris) ; 26(6-7): 647-51, 2010.
Article in French | MEDLINE | ID: mdl-20619169

ABSTRACT

Mental illnesses such as bipolar disorders, ADHD, depression and schizophrenia are a major public health concern worldwide. While several pharmacological agents acting on monoamine neurotransmission are used for the management of these disorders, the ultimate molecular mechanisms responsible for their therapeutic effects and their relationships to disease etiology are still poorly understood. Here we provide an overview of recent advances on the involvement of the signaling molecules Akt and glycogen synthase kinase-3 (GSK3) in the regulation of behavior by the monoamine neurotransmitters dopamine and serotonin (5-HT). We also examine the possible participation of these signaling molecules to the effects of antipsychotics, antidepressant and lithium and their contribution to mental disorders. Regulation of Akt and GSK3 by monoamine neurotransmitters may provide a better understanding of mental illnesses leading to new therapeutic approaches having better efficacy.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Mental Disorders/drug therapy , Proto-Oncogene Proteins c-akt/physiology , Psychotropic Drugs/therapeutic use , Signal Transduction/physiology , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Dopamine/pharmacology , Dopamine/therapeutic use , Glycogen Synthase Kinase 3/drug effects , Humans , Mental Disorders/physiopathology , Proto-Oncogene Proteins c-akt/drug effects , Psychotropic Drugs/pharmacology , Serotonin/pharmacology , Serotonin/therapeutic use , Signal Transduction/drug effects
12.
Behav Brain Res ; 205(1): 226-33, 2009 Dec 14.
Article in English | MEDLINE | ID: mdl-19683547

ABSTRACT

The delayed reaction paradigm, consisting to discover two different rules consecutively (delayed alternation and non-alternation task) followed by a delayed reversal task, is a specific marker for the functioning of primate prefrontal cortex. Although several works in rodents report the use of operant delayed alternation tasks, in none of the studies mice with lesion of the prefrontal cortex were used in this paradigm. In the current study, mouse experiments were conducted using a new, totally automated device, the olfactory H-maze. Here, we show that unilateral lesion of the dorsomedial prefrontal cortex in mice induced similar deficits to those observed after frontal lesions in monkeys and humans. These pronounced learning deficits seem to come from difficulty elaborating a new rule and the inability to inhibit the previous rule, characterized by perseveration after prefrontal cortex lesion. The present results demonstrate that this very simple experimental paradigm using the olfactory H-maze presents the advantage to be fast (one training session) and well suited to assess the frontal functions in mice. It should be useful for testing pharmacological or stem cell approaches in order to reduce organic damages or gain insight into the cognitive functions of the frontal cortex using transgenic or gene-targeting mice.


Subject(s)
Learning Disabilities/physiopathology , Maze Learning/physiology , Olfactory Perception/physiology , Prefrontal Cortex/physiopathology , Analysis of Variance , Animals , Automation , Learning Disabilities/etiology , Learning Disabilities/pathology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred BALB C , Neuropsychological Tests , Prefrontal Cortex/injuries , Prefrontal Cortex/pathology , Reversal Learning/physiology , Time Factors
13.
Mol Pharmacol ; 73(5): 1339-42, 2008 May.
Article in English | MEDLINE | ID: mdl-18314495

ABSTRACT

Antipsychotic agents are major drugs for human neuropsychiatric conditions including schizophrenia, mood disorders, Tourette syndrome, and Alzheimer's disease. These drugs are divided in two groups-first-generation/typical and second-generation/atypical-on the basis of their propensity to induce extrapyramidal motor side effects. Furthermore, second-generation antipsychotics have been reported to be superior in addressing cognitive deficits in schizophrenia. Understanding differences between the mechanism of action of first- and second-generation antipsychotic agents thus represents an interesting opportunity for the development of new compounds having better therapeutic action and less side effects. In this issue of Molecular Pharmacology, Fumagalli et al. (p. 1484) report that long-term treatment with the first-generation drug haloperidol interferes with the trafficking of both alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and N-methyl-D-aspartate glutamate receptor complexes and associated molecules post-synaptic densities 95 and Ca(2+)calmodulin-dependent protein kinase in the rat frontal cortex. In contrast, the second-generation drug olanzapine did not affect glutamate receptor trafficking. The action of haloperidol on glutamate receptor trafficking in specific brain regions may contribute to the low efficacy of this drug on cognitive deficits and to the development of side effects. Overall, antipsychotics have been shown to act upon multiple signaling mechanisms (e.g., cAMP-protein kinase A, betaArrestin 2-Akt-GSK-3, and phospholipase C-inositol-protein kinase C pathways), mostly by blocking D2-class dopamine receptors (first generation) or D2-class dopamine and 5-HT(2) serotonin receptors (second generation). Identification of specific pathways by which haloperidol affects glutamate receptor trafficking may thus represent an important next step toward the development of better antipsychotic drugs.


Subject(s)
Antipsychotic Agents/pharmacology , Receptors, Glutamate/metabolism , Animals , Antipsychotic Agents/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/enzymology , Prefrontal Cortex/metabolism , Protein Transport/drug effects , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL