Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Proc Natl Acad Sci U S A ; 119(37): e2120079119, 2022 09 13.
Article En | MEDLINE | ID: mdl-36067316

The extracellular protein Reelin, expressed by Cajal-Retzius (CR) cells at early stages of cortical development and at late stages by GABAergic interneurons, regulates radial migration and the "inside-out" pattern of positioning. Current models of Reelin functions in corticogenesis focus on early CR cell-derived Reelin in layer I. However, developmental disorders linked to Reelin deficits, such as schizophrenia and autism, are related to GABAergic interneuron-derived Reelin, although its role in migration has not been established. Here we selectively inactivated the Reln gene in CR cells or GABAergic interneurons. We show that CR cells have a major role in the inside-out order of migration, while CR and GABAergic cells sequentially cooperate to prevent invasion of cortical neurons into layer I. Furthermore, GABAergic cell-derived Reelin compensates some features of the reeler phenotype and is needed for the fine tuning of the layer-specific distribution of cortical neurons. In the hippocampus, the inactivation of Reelin in CR cells causes dramatic alterations in the dentate gyrus and mild defects in the hippocampus proper. These findings lead to a model in which both CR and GABAergic cell-derived Reelin cooperate to build the inside-out order of corticogenesis, which might provide a better understanding of the mechanisms involved in the pathogenesis of neuropsychiatric disorders linked to abnormal migration and Reelin deficits.


Cerebral Cortex , Nerve Tissue Proteins , Neurons , Reelin Protein , Animals , Cell Movement , Cerebral Cortex/cytology , Cerebral Cortex/embryology , GABAergic Neurons/enzymology , Hippocampus/embryology , Hippocampus/enzymology , Interneurons/enzymology , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/enzymology , Reelin Protein/genetics , Reelin Protein/metabolism
2.
Mol Brain ; 14(1): 139, 2021 09 10.
Article En | MEDLINE | ID: mdl-34507588

Chronic pain easily leads to concomitant mood disorders, and the excitability of anterior cingulate cortex (ACC) pyramidal neurons (PNs) is involved in chronic pain-related anxiety. However, the mechanism by which PNs regulate pain-related anxiety is still unknown. The GABAergic system plays an important role in modulating neuronal activity. In this paper, we aimed to study how the GABAergic system participates in regulating the excitability of ACC PNs, consequently affecting chronic inflammatory pain-related anxiety. A rat model of CFA-induced chronic inflammatory pain displayed anxiety-like behaviors, increased the excitability of ACC PNs, and reduced inhibitory presynaptic transmission; however, the number of GAD65/67 was not altered. Interestingly, intra-ACC injection of the GABAAR agonist muscimol relieved anxiety-like behaviors but had no effect on chronic inflammatory pain. Intra-ACC injection of the GABAAR antagonist picrotoxin induced anxiety-like behaviors but had no effect on pain in normal rats. Notably, chemogenetic activation of GABAergic neurons in the ACC alleviated chronic inflammatory pain and pain-induced anxiety-like behaviors, enhanced inhibitory presynaptic transmission, and reduced the excitability of ACC PNs. Chemogenetic inhibition of GABAergic neurons in the ACC led to pain-induced anxiety-like behaviors, reduced inhibitory presynaptic transmission, and enhanced the excitability of ACC PNs but had no effect on pain in normal rats. We demonstrate that the GABAergic system mediates a reduction in inhibitory presynaptic transmission in the ACC, which leads to enhanced excitability of pyramidal neurons in the ACC and is associated with chronic inflammatory pain-related anxiety.


Anxiety/physiopathology , Chronic Pain/physiopathology , GABAergic Neurons/physiology , Gyrus Cinguli/physiopathology , Inflammation/psychology , Pyramidal Cells/physiology , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Anxiety/etiology , Central Nervous System Sensitization/drug effects , Chronic Pain/psychology , Clozapine/therapeutic use , Freund's Adjuvant/toxicity , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Agonists/therapeutic use , GABA-A Receptor Antagonists/administration & dosage , GABA-A Receptor Antagonists/pharmacology , GABA-A Receptor Antagonists/toxicity , GABAergic Neurons/enzymology , Genetic Vectors/pharmacology , Inflammation/chemically induced , Inflammation/physiopathology , Injections , Interneurons/drug effects , Male , Muscimol/administration & dosage , Muscimol/pharmacology , Muscimol/therapeutic use , Open Field Test , Pain Threshold/drug effects , Patch-Clamp Techniques , Picrotoxin/toxicity , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Pyramidal Cells/enzymology , Rats , Rats, Sprague-Dawley
3.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Article En | MEDLINE | ID: mdl-34389674

Astrocytes have emerged as a potential source for new neurons in the adult mammalian brain. In mice, adult striatal neurogenesis can be stimulated by local damage, which recruits striatal astrocytes into a neurogenic program by suppression of active Notch signaling (J. P. Magnusson et al., Science 346, 237-241 [2014]). Here, we induced adult striatal neurogenesis in the intact mouse brain by the inhibition of Notch signaling in astrocytes. We show that most striatal astrocyte-derived neurons are confined to the anterior medial striatum, do not express established striatal neuronal markers, and exhibit dendritic spines, which are atypical for striatal interneurons. In contrast to striatal neurons generated during development, which are GABAergic or cholinergic, most adult astrocyte-derived striatal neurons possess distinct electrophysiological properties, constituting the only glutamatergic striatal population. Astrocyte-derived neurons integrate into the adult striatal microcircuitry, both receiving and providing synaptic input. The glutamatergic nature of these neurons has the potential to provide excitatory input to the striatal circuitry and may represent an efficient strategy to compensate for reduced neuronal activity caused by aging or lesion-induced neuronal loss.


Astrocytes/physiology , Connexin 30/metabolism , Glutamic Acid/metabolism , Neurons/physiology , Animals , Cell Differentiation , Connexin 30/genetics , Deoxyuridine/analogs & derivatives , Deoxyuridine/pharmacology , Electrophysiological Phenomena , GABAergic Neurons/enzymology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Interneurons/enzymology , Luminescent Proteins , Mice , Mice, Transgenic , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Recombination, Genetic , Tamoxifen/pharmacology
4.
Nature ; 590(7846): 451-456, 2021 02.
Article En | MEDLINE | ID: mdl-33361810

Reinforcement learning models postulate that neurons that release dopamine encode information about action and action outcome, and provide a teaching signal to striatal spiny projection neurons in the form of dopamine release1. Dopamine is thought to guide learning via dynamic and differential modulation of protein kinase A (PKA) in each class of spiny projection neuron2. However, the real-time relationship between dopamine and PKA in spiny projection neurons remains untested in behaving animals. Here we monitor the activity of dopamine-releasing neurons, extracellular levels of dopamine and net PKA activity in spiny projection neurons in the nucleus accumbens of mice during learning. We find positive and negative modulation of dopamine that evolves across training and is both necessary and sufficient to explain concurrent fluctuations in the PKA activity of spiny projection neurons. Modulations of PKA in spiny projection neurons that express type-1 and type-2 dopamine receptors are dichotomous, such that these neurons are selectively sensitive to increases and decreases, respectively, in dopamine that occur at different phases of learning. Thus, PKA-dependent pathways in each class of spiny projection neuron are asynchronously engaged by positive or negative dopamine signals during learning.


Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine/metabolism , Learning , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/metabolism , Female , Fluorescence , GABAergic Neurons/drug effects , GABAergic Neurons/enzymology , GABAergic Neurons/metabolism , Learning/drug effects , Male , Mice , Neuronal Plasticity/drug effects , Nucleus Accumbens/cytology , Photometry , Receptors, Dopamine/classification , Receptors, Dopamine/metabolism
5.
J Neurosci ; 40(11): 2332-2342, 2020 03 11.
Article En | MEDLINE | ID: mdl-32005763

Emotional disorders are common comorbid conditions that further exacerbate the severity and chronicity of chronic pain. However, individuals show considerable vulnerability to the development of chronic pain under similar pain conditions. In this study on male rat and mouse models of chronic neuropathic pain, we identify the histone deacetylase Sirtuin 1 (SIRT1) in central amygdala as a key epigenetic regulator that controls the development of comorbid emotional disorders underlying the individual vulnerability to chronic pain. We found that animals that were vulnerable to developing behaviors of anxiety and depression under the pain condition displayed reduced SIRT1 protein levels in central amygdala, but not those animals resistant to the emotional disorders. Viral overexpression of local SIRT1 reversed this vulnerability, but viral knockdown of local SIRT1 mimicked the pain effect, eliciting the pain vulnerability in pain-free animals. The SIRT1 action was associated with CaMKIIα downregulation and deacetylation of histone H3 lysine 9 at the CaMKIIα promoter. These results suggest that, by transcriptional repression of CaMKIIα in central amygdala, SIRT1 functions to guard against the emotional pain vulnerability under chronic pain conditions. This study indicates that SIRT1 may serve as a potential therapeutic molecule for individualized treatment of chronic pain with vulnerable emotional disorders.SIGNIFICANCE STATEMENT Chronic pain is a prevalent neurological disease with no effective treatment at present. Pain patients display considerably variable vulnerability to developing chronic pain, indicating individual-based molecular mechanisms underlying the pain vulnerability, which is hardly addressed in current preclinical research. In this study, we have identified the histone deacetylase Sirtuin 1 (SIRT1) as a key regulator that controls this pain vulnerability. This study reveals that the SIRT1-CaMKIIaα pathway in central amygdala acts as an epigenetic mechanism that guards against the development of comorbid emotional disorders under chronic pain, and that its dysfunction causes increased vulnerability to the development of chronic pain. These findings suggest that SIRT1 activators may be used in a novel therapeutic approach for individual-based treatment of chronic pain.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Central Amygdaloid Nucleus/physiopathology , Chronic Pain/physiopathology , Psychological Distress , Sirtuin 1/physiology , Trigeminal Neuralgia/physiopathology , Acetylation , Animals , Anxiety/etiology , Anxiety/physiopathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Central Amygdaloid Nucleus/enzymology , Chronic Pain/psychology , Depression/etiology , Depression/physiopathology , Disease Susceptibility , Down-Regulation , Exploratory Behavior , GABAergic Neurons/enzymology , Genetic Vectors , Histones/metabolism , Hyperalgesia/physiopathology , Male , Mice , Promoter Regions, Genetic , Rats , Rats, Wistar , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics , Swimming , Transcription, Genetic , Trigeminal Neuralgia/psychology
6.
Elife ; 82019 10 28.
Article En | MEDLINE | ID: mdl-31657720

Parvalbumin-expressing (PV+) GABAergic interneurons mediate feedforward and feedback inhibition and have a key role in gamma oscillations and information processing. The importance of fast synaptic recruitment and action potential initiation and repolarization, and rapid synchronous GABA release by PV+ cells, is well established. In contrast, the functional significance of PV+ cell NMDA receptors (NMDARs), which generate relatively slow postsynaptic currents, is unclear. Underlining their potential importance, several studies implicate PV+ cell NMDAR disruption in impaired network function and circuit pathologies. Here, we show that dendritic NMDARs underlie supralinear integration of feedback excitation from local pyramidal neurons onto mouse CA1 PV+ cells. Furthermore, by incorporating NMDARs at feedback connections onto PV+ cells in spiking networks, we show that these receptors enable cooperative recruitment of PV+ interneurons, strengthening and stabilising principal cell assemblies. Failure of this phenomenon provides a parsimonious explanation for cognitive and sensory gating deficits in pathologies with impaired PV+ NMDAR signalling.


Dendrites/physiology , GABAergic Neurons/physiology , Interneurons/physiology , Nerve Net/physiology , Pyramidal Cells/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Action Potentials , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Dendrites/enzymology , GABAergic Neurons/enzymology , Interneurons/enzymology , Mice
7.
J Psychiatr Res ; 115: 21-28, 2019 08.
Article En | MEDLINE | ID: mdl-31082653

Na+, K+-ATPase is an essential membrane transporter. In the brain, the α3 isoform of Na+, K+-ATPase is vital for neuronal function. The enzyme and its regulators, endogenous cardiac steroids (ECS), were implicated in neuropsychiatric disorders. GABAergic neurotransmission was also studied extensively in diseases such as schizophrenia and bipolar disorder (BD). Post mortem brain samples from subjects with depression, schizophrenia or BD and non-psychiatric controls were provided by the Stanley Medical Research Institute. ECS levels were determined by ELISA. Expression levels of the three Na+, K+-ATPase-α isoforms, α1, α2 and α3, were determined by Western blot analysis. The α3 levels in GABAergic neurons in different regions of the brain were quantified by fluorescence immunohistochemistry. The results show that Na+, K+ -ATPase α3 isoform levels were lower in GABAergic neurons in the frontal cortex in BD and schizophrenia as compared with the controls (n = 15 subjects per group). A study on a 'mini-cohort' (n = 3 subjects per group) showed that the α3 isoform levels were also lower in GABAergic neurons in the hippocampus, but not amygdala, of bipolar and schizophrenic subjects. In the temporal cortex, higher Na+, K+ -ATPase α3 protein levels were found in the three psychiatric groups. No significant differences in ECS levels were found in this brain area. This is the first report on the distribution of α3 in specific neurons in the human brain in association with mental illness. These results strengthen the hypothesis for the involvement of Na+, K+ -ATPase in neuropsychiatric diseases.


Bipolar Disorder/enzymology , Depressive Disorder/enzymology , GABAergic Neurons/enzymology , Interneurons/enzymology , Prefrontal Cortex/enzymology , Schizophrenia/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Tissue Banks , Adult , Amygdala/enzymology , Hippocampus/enzymology , Humans , Prefrontal Cortex/pathology , Protein Isoforms , Temporal Lobe/enzymology
8.
Brain Struct Funct ; 224(1): 453-469, 2019 Jan.
Article En | MEDLINE | ID: mdl-30368554

In mammals, the extended amygdala is a neural hub for social and emotional information processing. In the rat, the extended amygdala receives inhibitory GABAergic projections from the nucleus incertus (NI) in the pontine tegmentum. NI neurons produce the neuropeptide relaxin-3, which acts via the Gi/o-protein-coupled receptor, RXFP3. A putative role for RXFP3 signalling in regulating social interaction was investigated by assessing the effect of intracerebroventricular infusion of the RXFP3 agonist, RXFP3-A2, on performance in the 3-chamber social interaction paradigm. Central RXFP3-A2, but not vehicle, infusion, disrupted the capacity to discriminate between a familiar and novel conspecific subject, but did not alter differentiation between a conspecific and an inanimate object. Subsequent studies revealed that agonist-infused rats displayed increased phosphoERK(pERK)-immunoreactivity in specific amygdaloid nuclei at 20 min post-infusion, with levels similar to control again after 90 min. In parallel, we used immunoblotting to profile ERK phosphorylation dynamics in whole amygdala after RXFP3-A2 treatment; and multiplex histochemical labelling techniques to reveal that after RXFP3-A2 infusion and social interaction, pERK-immunopositive neurons in amygdala expressed vesicular GABA-transporter mRNA and displayed differential profiles of RXFP3 and oxytocin receptor mRNA. Overall, these findings demonstrate that central relaxin-3/RXFP3 signalling can modulate social recognition in rats via effects within the amygdala and likely interactions with GABA and oxytocin signalling.


Amygdala/drug effects , Behavior, Animal/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , GABAergic Neurons/drug effects , Peptides/administration & dosage , Receptors, G-Protein-Coupled/agonists , Receptors, Peptide/agonists , Recognition, Psychology/drug effects , Social Behavior , gamma-Aminobutyric Acid/metabolism , Amygdala/cytology , Amygdala/enzymology , Animals , GABAergic Neurons/enzymology , Infusions, Intraventricular , Intercellular Signaling Peptides and Proteins , Male , Oxytocin/metabolism , Phosphorylation , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Signal Transduction/drug effects , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
9.
Transl Psychiatry ; 8(1): 271, 2018 12 10.
Article En | MEDLINE | ID: mdl-30531788

Experience of traumatic events in childhood is linked to an elevated risk of developing psychiatric disorders in adulthood. The neurobiological mechanisms underlying this phenomenon are not fully understood. The limbic system, particularly the hippocampus, is significantly impacted by childhood trauma. In particular, it has been hypothesised that childhood stress may impact adult hippocampal neurogenesis (AHN) and related behaviours, conferring increased risk for later mental illness. Stress in utero can lead to impaired hippocampal synaptic plasticity, and stress in the first 2-3 weeks of life reduces AHN in animal models. Less is known about the effects of stress in the post-weaning, pre-pubertal phase, a developmental time-point more akin to human childhood. Therefore, we investigated persistent effects of pre-pubertal stress (PPS) on functional and molecular aspects of the hippocampus. AHN was altered following PPS in male rats only. Specifically males showed reduced production of new neurons following PPS, but increased survival in the ventral dentate gyrus. In adult males, but not females, pattern separation and trace fear conditioning, behaviours that rely heavily on AHN, were also impaired after PPS. PPS also increased the expression of parvalbumin-positive GABAergic interneurons in the ventral dentate gyrus and increased glutamic acid decarboxylase 67 expression in the ventral hilus, in males only. Our results demonstrate the lasting effects of PPS on the hippocampus in a sex- and time-dependent manner, provide a potential mechanistic link between PPS and later behavioural impairments, and highlight sex differences in vulnerability to neuropsychiatric conditions after early-life stress.


Behavior, Animal , Hippocampus/physiopathology , Neurogenesis , Sex Characteristics , Stress, Psychological/physiopathology , Animals , Conditioning, Classical , Discrimination, Psychological , Fear , Female , GABAergic Neurons/enzymology , Glutamate Decarboxylase/metabolism , Interneurons/enzymology , Male , Pattern Recognition, Physiological , Puberty , Rats
10.
Proc Natl Acad Sci U S A ; 115(10): E2437-E2446, 2018 03 06.
Article En | MEDLINE | ID: mdl-29463694

Circadian disruption as a result of shift work is associated with adverse metabolic consequences. Internal desynchrony between the phase of the suprachiasmatic nuclei (SCN) and peripheral clocks is widely believed to be a major factor contributing to these adverse consequences, but this hypothesis has never been tested directly. A GABAergic Cre driver combined with conditional casein kinase mutations (Vgat-Cre+CK1δfl/flεfl/+ ) was used to lengthen the endogenous circadian period in GABAergic neurons, including the SCN, but not in peripheral tissues, to create a Discordant mouse model. These mice had a long (27.4 h) behavioral period to which peripheral clocks entrained in vivo, albeit with an advanced phase (∼6 h). Thus, in the absence of environmental timing cues, these mice had internal desynchrony between the SCN and peripheral clocks. Surprisingly, internal desynchrony did not result in obesity in this model. Instead, Discordant mice had reduced body mass compared with Cre-negative controls on regular chow and even when challenged with a high-fat diet. Similarly, internal desynchrony failed to induce glucose intolerance or disrupt body temperature and energy expenditure rhythms. Subsequently, a lighting cycle of 2-h light/23.5-h dark was used to create a similar internal desynchrony state in both genotypes. Under these conditions, Discordant mice maintained their lower body mass relative to controls, suggesting that internal desynchrony did not cause the lowered body mass. Overall, our results indicate that internal desynchrony does not necessarily lead to metabolic derangements and suggest that additional mechanisms contribute to the adverse metabolic consequences observed in circadian disruption protocols.


Casein Kinase 1 epsilon/genetics , Casein Kinase Idelta/genetics , Circadian Clocks , GABAergic Neurons/enzymology , Suprachiasmatic Nucleus/physiology , Animals , Casein Kinase 1 epsilon/deficiency , Casein Kinase Idelta/deficiency , Circadian Rhythm , Female , Gene Knockout Techniques , Gene Silencing , Male , Mice , Mice, Inbred C57BL , Suprachiasmatic Nucleus/enzymology
11.
Cereb Cortex ; 27(12): 5696-5714, 2017 12 01.
Article En | MEDLINE | ID: mdl-29117290

The proliferative niches in the subpallium generate a rich cellular variety fated for diverse telencephalic regions. The embryonic preoptic area (POA) represents one of these domains giving rise to the pool of cortical GABAergic interneurons and glial cells, in addition to striatal and residual POA cells. The migration from sites of origin within the subpallium to the distant targets like the cerebral cortex, accomplished by the adoption and maintenance of a particular migratory morphology, is a critical step during interneuron development. To identify factors orchestrating this process, we performed single-cell transcriptome analysis and detected Dnmt1 expression in murine migratory GABAergic POA-derived cells. Deletion of Dnmt1 in postmitotic immature cells of the POA caused defective migration and severely diminished adult cortical interneuron numbers. We found that DNA methyltransferase 1 (DNMT1) preserves the migratory shape in part through negative regulation of Pak6, which stimulates neuritogenesis at postmigratory stages. Our data underline the importance of DNMT1 for the migration of POA-derived cells including cortical interneurons.


Cell Movement/physiology , Cerebral Cortex/embryology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Interneurons/enzymology , Neural Stem Cells/enzymology , Preoptic Area/embryology , Animals , Animals, Newborn , Cell Count , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , DNA Methylation , GABAergic Neurons/cytology , GABAergic Neurons/enzymology , Interneurons/cytology , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/cytology , Neuronal Outgrowth/physiology , Preoptic Area/cytology , Preoptic Area/enzymology , Tissue Culture Techniques , Transcriptome , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
12.
Neurosci Lett ; 649: 48-54, 2017 05 10.
Article En | MEDLINE | ID: mdl-28400125

Dysfunction of inhibitory GABAergic interneurons is considered a major pathophysiological feature of various neurodevelopmental and neuropsychiatric disorders. The variants of SHANK3 gene, encoding a core scaffold protein of the excitatory postsynapse, have been associated with numerous brain disorders. It has been suggested that abnormalities of GABAergic interneurons could contribute to the SHANK3-related disorders, but the limitation of these studies is that they used mainly Shank3 knock-out mice. Notably, Shank3-overexpressing transgenic mice, modeling human hyperkinetic disorders, also show reduced inhibitory synaptic transmission, abnormal electroencephalography, and spontaneous seizures. However, it has not been investigated whether these phenotypes of Shank3 transgenic mice are associated with GABAergic interneuron dysfunction, or solely due to the cell-autonomous postsynaptic changes of principal neurons. To address this issue, we investigated the densities of parvalbumin- and somatostatin-positive interneurons, and the mRNA and protein levels of GAD65/67 GABA-synthesizing enzymes in the medial prefrontal cortex, striatum, and hippocampus of adult Shank3 transgenic mice. We found no significant difference in the measurements performed on wild-type versus Shank3 transgenic mice, except for the decreased GAD65 or GAD67 mRNAs in these brain regions. Interestingly, only GAD65 mRNA was decreased in the hippocampus, but not mPFC and striatum, of juvenile Shank3 transgenic mice which, unlike the adult mice, did not show behavioral hyperactivity. Together, our results suggest age-dependent decrease of GAD65/67 mRNAs but normal densities of certain GABAergic interneurons in the Shank3 transgenic mice.


Bipolar Disorder/enzymology , Brain/enzymology , GABAergic Neurons/enzymology , Glutamate Decarboxylase/metabolism , Nerve Tissue Proteins/metabolism , Animals , Corpus Striatum/enzymology , Disease Models, Animal , Hippocampus/enzymology , Interneurons/enzymology , Male , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins , Parvalbumins/metabolism , Prefrontal Cortex/enzymology , RNA, Messenger/metabolism , Somatostatin/metabolism
13.
Oncotarget ; 8(24): 39309-39322, 2017 Jun 13.
Article En | MEDLINE | ID: mdl-28445148

Cerebral ischemia leads to neuronal death for stroke, in which the imbalance between glutamatergic neurons and GABAergic neurons toward neural excitotoxicity is presumably involved. GABAergic neurons are vulnerable to pathological factors and impaired in an early stage of ischemia. The rescue of GABAergic neurons is expected to be the strategy to reserve ischemic neuronal impairment. As protein kinase C (PKC) and calmodulin-dependent protein kinase II (CaMK-II) are activated during ischemia, we have investigated whether the inhibitions of these kinases rescue the ischemic impairment of cortical GABAergic neurons. The functions of GABAergic neurons were analyzed by whole-cell recording in the cortical slices during ischemia and in presence of 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-L-tyrosyl]-4-phenylpiperazine (CaMK-II inhibitor) and chelerythrine chloride (PKC inhibitor). Our results indicate that PKC inhibitor or CaMK-II inhibitor partially prevents ischemia-induced functional deficits of cortical GABAergic neurons. Moreover, the combination of PKC and CaMK-II inhibitors synergistically reverses this ischemia-induced deficit of GABAergic neurons. One of potential therapeutic strategies for ischemic stroke may be to rescue the ischemia-induced deficit of cortical GABAergic neurons by inhibiting PKC and CaMK-II.


Benzophenanthridines/pharmacology , Brain Ischemia/complications , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , GABAergic Neurons/drug effects , Protein Kinase C/antagonists & inhibitors , Stroke/prevention & control , Animals , Antineoplastic Agents/pharmacology , Brain Ischemia/physiopathology , Cells, Cultured , GABAergic Neurons/enzymology , GABAergic Neurons/pathology , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Stroke/enzymology , Stroke/etiology , Stroke/pathology
14.
Exp Neurol ; 287(Pt 1): 44-53, 2017 Jan.
Article En | MEDLINE | ID: mdl-27780732

GABAergic disinhibition has been suggested to play a critical role in the pathophysiology of several basal ganglia disorders, including dystonia, a common movement disorder. Previous studies have shown a deficit of striatal GABAergic interneurons (IN) in the dtsz mutant hamster, one of the few phenotypic animal models of dystonia. However, mechanisms underlying this deficit are largely unknown. In the present study, we investigated the migration and maturation of striatal IN during postnatal development (18days of age) and at age of highest severity of dystonia (33days of age) in this hamster model. In line with previous findings, the density of GAD67-positive IN and the level of parvalbumin mRNA, a marker for fast spiking GABAergic IN, were lower in the dtsz mutant than in control hamsters. However, an unaltered density of Nkx2.1 labeled cells and Nkx2.1 mRNA level suggested that the migration of GABAergic IN into the striatum was not retarded. Therefore, different factors that indicate maturation of GABAergic IN were determined. While mRNA of the KCC2 cation/chloride transporters and the cytosolic carboanhydrase VII, used as markers for the so called GABA switch, as well as BDNF were unaltered, we found a reduced number of IN expressing the alpha1 subunit of the GABAA-receptor (37.5%) in dtsz hamsters at an age of 33days, but not after spontaneous remission of dystonia at an age of 90days. Since IN shift expression from alpha2 to alpha1 subunits during postnatal maturation, this result together with a decreased parvalbumin mRNA expression suggest a delayed maturation of striatal GABAergic IN in this animal model, which might underlie abnormal neuronal activity and striatal plasticity.


Corpus Striatum/growth & development , Corpus Striatum/pathology , Dystonia/pathology , GABAergic Neurons/physiology , Gene Expression Regulation, Developmental/physiology , Action Potentials/drug effects , Action Potentials/genetics , Age Factors , Animals , Animals, Newborn , Cricetinae , Disease Models, Animal , Dystonia/genetics , Female , GABAergic Neurons/enzymology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/genetics , Glutamate Decarboxylase/metabolism , Male , Mesocricetus/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Parvalbumins/genetics , Parvalbumins/metabolism , Phenotype , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Symporters/genetics , Symporters/metabolism , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/metabolism , K Cl- Cotransporters
15.
Pharmacogenomics ; 18(1): 17-22, 2017 Jan.
Article En | MEDLINE | ID: mdl-27967329

AIM: Association between polymorphisms in GAD genes and methamphetamine (METH) dependence was investigated in the Thai population. MATERIALS & METHODS: Genotypes of rs769404 and rs701492 in GAD1 and rs2236418 in GAD2 polymorphisms were determined in 100 METH-dependent male subjects and 102 matched controls. RESULTS: The genotype and allele frequencies of rs2236418 (GAD2) were associated with METH dependence and METH with psychosis, in which the G allele was related to increased risk. The presence of the rs769404-rs701492 (GAD1) C-C haplotype was associated with METH psychosis. CONCLUSION: This study indicates that genetic variability in GAD1 and GAD2 contributes to risk of METH dependence and METH psychosis in the Thai population and indicates the role of the GABAergic system in these disorders.


Amphetamine-Related Disorders/genetics , Glutamate Decarboxylase/genetics , Methamphetamine , Polymorphism, Single Nucleotide/genetics , Adult , Amphetamine-Related Disorders/diagnosis , Amphetamine-Related Disorders/epidemiology , Cohort Studies , GABAergic Neurons/enzymology , Humans , Male , Middle Aged , Thailand/epidemiology , Young Adult
16.
Neurobiol Aging ; 48: 83-92, 2016 12.
Article En | MEDLINE | ID: mdl-27644077

Impaired clearance of amyloid-ß peptide (Aß) has been postulated to significantly contribute to the amyloid accumulation typical of Alzheimer's disease. Among the enzymes known to degrade Aß in vivo are endothelin-converting enzyme (ECE)-1, ECE-2, and neprilysin (NEP), and evidence suggests that they regulate independent pools of Aß that may be functionally significant. To better understand the differential regulation of Aß concentration by its physiological degrading enzymes, we characterized the cell and region-specific expression pattern of ECE-1, ECE-2, and NEP by in situ hybridization and immunohistochemistry in brain areas relevant to Alzheimer's disease. In contrast to the broader distribution of ECE-1, ECE-2 and NEP were found enriched in GABAergic neurons. ECE-2 was majorly expressed by somatostatin-expressing interneurons and was active in isolated synaptosomes. NEP messenger RNA was found mainly in parvalbumin-expressing interneurons, with NEP protein localized to perisomatic parvalbuminergic synapses. The identification of somatostatinergic and parvalbuminergic synapses as hubs for Aß degradation is consistent with the possibility that Aß may have a physiological function related to the regulation of inhibitory signaling.


Amyloid beta-Peptides/metabolism , Endothelin-Converting Enzymes/metabolism , GABAergic Neurons/enzymology , Hippocampus/cytology , Hippocampus/enzymology , Neocortex/cytology , Neocortex/enzymology , Neprilysin/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/physiology , Animals , Endothelin-Converting Enzymes/genetics , Endothelin-Converting Enzymes/physiology , Gene Expression , Mice, Transgenic , Neprilysin/genetics , Neprilysin/physiology , RNA, Messenger/metabolism , Synapses/enzymology
17.
Anat Sci Int ; 91(4): 398-406, 2016 Sep.
Article En | MEDLINE | ID: mdl-26643381

The septum is a basal forebrain region located between the lateral ventricles in rodents. It consists of lateral and medial divisions. Medial septal projections regulate hippocampal theta rhythm whereas lateral septal projections are involved in processes such as affective functions, memory formation, and behavioral responses. Gamma-aminobutyric acidergic neurons of the septal region possess the 65 and 67 isoforms of the enzyme glutamic acid decarboxylase. Although data on the glutamic acid decarboxylase isoform distribution in the septal region generally appears to indicate glutamic acid decarboxylase 67 dominance, different studies have given inconsistent results in this regard. The aim of this study was therefore to obtain information on the distributions of both of these glutamic acid decarboxylase isoforms in the septal region in transgenic mice. Two animal groups of glutamic acid decarboxylase-green fluorescent protein knock-in transgenic mice were utilized in the experiment. Brain sections from the region were taken for anti-green fluorescent protein immunohistochemistry in order to obtain estimated quantitative data on the number of gamma-aminobutyric acidergic neurons. Following the immunohistochemical procedures, the mean numbers of labeled cells in the lateral and medial septal nuclei were obtained for the two isoform groups. Statistical analysis yielded significant results which indicated that the 65 isoform of glutamic acid decarboxylase predominates in both lateral and medial septal nuclei (unpaired two-tailed t-test p < 0.0001 for LS, p < 0.01 for MS). This study is the first to reveal the dominance of glutamic acid decarboxylase isoform 65 in the septal region in glutamic acid decarboxylase-green fluorescent protein transgenic mice.


Glutamate Decarboxylase/metabolism , Mice, Transgenic , Septum of Brain/enzymology , Animals , GABAergic Neurons/enzymology , GABAergic Neurons/physiology , Green Fluorescent Proteins , Immunohistochemistry/methods , Isoenzymes/metabolism , Septum of Brain/cytology , Septum of Brain/physiology
18.
Cereb Cortex ; 26(5): 2191-2204, 2016 May.
Article En | MEDLINE | ID: mdl-25824535

Non-overlapping groups of cortical γ-aminobutyric acid-releasing (GABAergic) neurons are identifiable by the presence of calbindin (CB), calretinin (CR), or parvalbumin (PV). Boutons from PV neuron subtypes are also distinguishable by differences in protein levels of the GABA-synthesizing enzymes GAD65 and GAD67. Multilabel fluorescence microscopy was used to determine if this diversity extends to boutons of CB and CR neurons in monkey prefrontal cortex. CB and CR neurons gave rise to 3 subpopulations of GAD-containing boutons: GAD65+, GAD67+, and GAD65/GAD67+. Somatostatin and vasoactive intestinal peptide-expressing neurons, subtypes of CB and CR neurons, respectively, also gave rise to these distinct bouton subpopulations. At the transcript level, CB and CR neurons contained mRNA encoding GAD67-only or both GADs. Thus, the distinct subpopulations of CB/GAD+ and CR/GAD+ boutons arise from 2 unique subtypes of CB and CR neurons. The different CB and CR GAD-expressing neurons targeted the same projection neurons and neuronal structures immunoreactive for PV, CR, or CB. These findings suggest that GABA synthesis from CB/GAD67+ and CR/GAD67+ neurons would presumably be more vulnerable to disease-associated deficits in GAD67 expression, such as in schizophrenia, than neurons that also contain GAD65.


Calbindin 2/metabolism , Calbindins/metabolism , GABAergic Neurons/enzymology , Prefrontal Cortex/enzymology , Presynaptic Terminals/enzymology , gamma-Aminobutyric Acid/biosynthesis , Animals , Glutamate Decarboxylase/metabolism , Macaca mulatta , Male , Somatostatin/metabolism , Vasoactive Intestinal Peptide/metabolism
19.
PLoS One ; 10(8): e0134296, 2015.
Article En | MEDLINE | ID: mdl-26241953

Homeostatic synaptic plasticity, or synaptic scaling, is a mechanism that tunes neuronal transmission to compensate for prolonged, excessive changes in neuronal activity. Both excitatory and inhibitory neurons undergo homeostatic changes based on synaptic transmission strength, which could effectively contribute to a fine-tuning of circuit activity. However, gene regulation that underlies homeostatic synaptic plasticity in GABAergic (GABA, gamma aminobutyric) neurons is still poorly understood. The present study demonstrated activity-dependent dynamic scaling in which NMDA-R (N-methyl-D-aspartic acid receptor) activity regulated the expression of GABA synthetic enzymes: glutamic acid decarboxylase 65 and 67 (GAD65 and GAD67). Results revealed that activity-regulated BDNF (brain-derived neurotrophic factor) release is necessary, but not sufficient, for activity-dependent up-scaling of these GAD isoforms. Bidirectional forms of activity-dependent GAD expression require both BDNF-dependent and BDNF-independent pathways, both triggered by NMDA-R activity. Additional results indicated that these two GAD genes differ in their responsiveness to chronic changes in neuronal activity, which could be partially caused by differential dependence on BDNF. In parallel to activity-dependent bidirectional scaling in GAD expression, the present study further observed that a chronic change in neuronal activity leads to an alteration in neurotransmitter release from GABAergic neurons in a homeostatic, bidirectional fashion. Therefore, the differential expression of GAD65 and 67 during prolonged changes in neuronal activity may be implicated in some aspects of bidirectional homeostatic plasticity within mature GABAergic presynapses.


Brain-Derived Neurotrophic Factor/physiology , Gene Expression Regulation , Glutamate Decarboxylase/biosynthesis , Signal Transduction/physiology , Animals , Benzylamines/pharmacology , Bicuculline/pharmacology , Butadienes/pharmacology , Calcium Signaling/drug effects , Carbazoles/pharmacology , Cells, Cultured , Cerebral Cortex/cytology , Enzyme Induction/drug effects , GABAergic Neurons/drug effects , GABAergic Neurons/enzymology , GABAergic Neurons/metabolism , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/genetics , Homeostasis , Indole Alkaloids/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred ICR , Nitriles/pharmacology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction/drug effects , Sulfonamides/pharmacology , gamma-Aminobutyric Acid/metabolism
20.
J Chem Neuroanat ; 61-62: 51-63, 2014 Nov.
Article En | MEDLINE | ID: mdl-25058170

As γ-aminobutyric acid (GABA) is synthesized by two isoforms of glutamic acid decarboxylase (GAD), namely, GAD65 and GAD67, immunohistochemically targeting either isoform of GAD is theoretically useful for identifying GABAergic cell bodies. In practice, targeting GAD67 remains to be a popular choice. However, identifying GABAergic cell bodies with GAD67 immunoreactivity in the hippocampal dentate gyrus, especially in the hilus, is not without pitfalls. In the present study, we compared the characteristics of GAD65 immunoreactivity to GAD67 immunoreactivity in the rat dentate gyrus and examined perikaryal expression of GAD65 in four neurochemically prevalent subgroups of interneurons in the hilus. Experiments were done in normal adult Sprague-Dawley rats and GAD67-GFP knock-in mice. Horizontal hippocampal slices cut from the ventral portion of hippocampi were immunofluorescently stained and scanned using a confocal microscope. Immunoreactivity for both GAD67 and GAD65 was visible throughout the dentate gyrus. Perikaryal GAD67 immunoreactivity was denser but variable in terms of distribution pattern and intensity among cells whereas perikaryal GAD65 immunoreactivity displayed similar distribution pattern and staining intensity. Among different layers of the dentate gyrus, GAD67 immunoreactivity was densest in the hilus despite GAD65 immunoreactivity being more intense in the granule cell layer. Co-localization experiments showed that GAD65, but not GAD67, was expressed in all hilar calretinin (CR)-, neuronal nitric oxide synthase (nNOS)-, parvalbumin (PV)- or somatostatin (SOM)-positive somata. Labeling CR, nNOS, PV, and SOM in sections obtained from GAD67-GFP knock-in mice revealed that a large portion of SOM-positive cells had weak GFP expression. In addition, double labeling of GAD65/GABA and GAD67/GABA showed that nearly all of GABA-immunoreactive cells had perikaryal GAD65 expression whereas more than one-tenth of GABA-immunoreactive cells lacked perikaryal GAD67 immunoreactivity. Inhibition of axonal transport with colchicine dramatically improved perikaryal GAD65 immunoreactivity in GABAergic cells without significant augmentation to be seen in granule cells. Double labeling GAD65 and GAD67 in the sections obtained from colchicine-pretreated animals confirmed that a portion of GAD65-immunoreactive cells had weak or even no GAD67 immunoreactivity. We conclude that for confocal imaging, immunofluorescently labeling GAD65 for identifying GABAergic somata in the hilus of the dentate gyrus has advantages over labeling GAD67 in terms of easier recognition of perikaryal labeling and more consistent expression in GABAergic somata. Inhibition of axonal transport with colchicine further improves perikaryal GAD65 labeling, making GABAergic cells more distinguishable.


Dentate Gyrus/cytology , GABAergic Neurons/cytology , Glutamate Decarboxylase/analysis , Animals , Dentate Gyrus/enzymology , Fluorescent Antibody Technique/methods , GABAergic Neurons/enzymology , Interneurons/cytology , Interneurons/enzymology , Male , Microscopy, Confocal , Rats , Rats, Sprague-Dawley
...