Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Brain Res Bull ; 205: 110830, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38036272

ABSTRACT

Premenstrual dysphoric disorder (PMDD) is a periodic psychiatric disorder with high prevalence in women of childbearing age, seriously affecting patients' work and life. Currently, the international first-line drugs for PMDD have low efficiency and increased side effects. Paeonol, a major component of the traditional Chinese medicine Cortex Moutan, has been applied in treating PMDD in China with satisfactory results, but the therapeutic mechanism is not fully understood. This study aims to evaluate the therapeutic effects and pharmacological mechanisms of paeonol on the main psychiatric symptoms and hippocampal damage in PMDD. We established a premenstrual irritability rat model by the resident-intruder paradigm and performed elevated plus maze and social interactions. And we employed the HE and Nissl staining techniques to observe the therapeutic effect of paeonol on hippocampal damage in PMDD rats. Subsequently, Elisa, qRT-PCR Array, Western Blotting, and cell models were utilized to elucidate the underlying molecular mechanisms through which paeonol intervenes in treating PMDD. In this study, we demonstrated the therapeutic effects of paeonol on irritability, anxiety, and social withdrawal behaviors in rats. In addition, we found that paeonol significantly reduced the serum corticosterone (CORT) level, improved hippocampal morphological structure and neuron number, and reduced hippocampal neuron apoptosis in PMDD rats. Paeonol reduced GRM5, GABBR2, ß-arrestin2, and GRK3 expression levels in hippocampal brain regions of PMDD rats and activated the cAMP/PKA signaling pathway. Inhibitor cell experiments showed that paeonol specifically ameliorated hippocampal injury by modulating the ß-arrestin2/PDE4-cAMP/PKA signaling pathway. The present study demonstrates, for the first time, that paeonol exerts a therapeutic effect on periodic psychotic symptoms and hippocampal injury in PMDD through inhibiting GRM5/GABBR2/ß-arrestin2 and activating cAMP-PKA signaling pathway. These findings enhance our understanding of the pharmacological mechanism underlying paeonol and provide a solid scientific foundation for its future clinical application.


Subject(s)
Premenstrual Dysphoric Disorder , Animals , Female , Rats , Acetophenones , Anxiety , Hippocampus/metabolism , Premenstrual Dysphoric Disorder/diagnosis , Premenstrual Dysphoric Disorder/epidemiology , Premenstrual Dysphoric Disorder/psychology , Receptors, GABA-B/metabolism
2.
Brain ; 145(7): 2332-2346, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35134125

ABSTRACT

Metabolism regulates neuronal activity and modulates the occurrence of epileptic seizures. Here, using two rodent models of absence epilepsy, we show that hypoglycaemia increases the occurrence of spike-wave seizures. We then show that selectively disrupting glycolysis in the thalamus, a structure implicated in absence epilepsy, is sufficient to increase spike-wave seizures. We propose that activation of thalamic AMP-activated protein kinase, a sensor of cellular energetic stress and potentiator of metabotropic GABAB-receptor function, is a significant driver of hypoglycaemia-induced spike-wave seizures. We show that AMP-activated protein kinase augments postsynaptic GABAB-receptor-mediated currents in thalamocortical neurons and strengthens epileptiform network activity evoked in thalamic brain slices. Selective thalamic AMP-activated protein kinase activation also increases spike-wave seizures. Finally, systemic administration of metformin, an AMP-activated protein kinase agonist and common diabetes treatment, profoundly increased spike-wave seizures. These results advance the decades-old observation that glucose metabolism regulates thalamocortical circuit excitability by demonstrating that AMP-activated protein kinase and GABAB-receptor cooperativity is sufficient to provoke spike-wave seizures.


Subject(s)
Epilepsy, Absence , Hypoglycemia , AMP-Activated Protein Kinases/metabolism , Epilepsy, Absence/metabolism , Humans , Hypoglycemia/chemically induced , Hypoglycemia/metabolism , Receptors, GABA-B/metabolism , Seizures , Thalamus
3.
Neuron ; 109(17): 2682-2690.e5, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34314698

ABSTRACT

Slow-wave sleep is characterized by near-synchronous alternation of active Up states and quiescent Down states in the neocortex. Although the cortex itself can maintain these oscillations, the full expression of Up-Down states requires intact thalamocortical circuits. Sensory thalamic input can drive the cortex into an Up state. Here we show that midline thalamic neurons terminate Up states synchronously across cortical areas. Combining local field potential, single-unit, and patch-clamp recordings in conjunction with optogenetic stimulation and silencing in mice in vivo, we report that thalamic input mediates Down transition via activation of layer 1 neurogliaform inhibitory neurons acting on GABAB receptors. These results strengthen the evidence that thalamocortical interactions are essential for the full expression of slow-wave sleep, show that Down transition is an active process mediated by cortical GABAB receptors, and demonstrate that thalamus synchronizes Down transitions across cortical areas during natural slow-wave sleep.


Subject(s)
Interneurons/physiology , Neocortex/physiology , Receptors, GABA-B/metabolism , Sleep, Slow-Wave/physiology , Thalamus/physiology , Animals , Evoked Potentials , Female , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/metabolism , Thalamus/cytology , Thalamus/metabolism
4.
Neurosci Lett ; 750: 135810, 2021 04 17.
Article in English | MEDLINE | ID: mdl-33705929

ABSTRACT

Although clinical efficacy of waggle needling has been confirmed, therapeutic mechanisms still remain poorly understood. Reduction of GABA was involved in the etiology of spasticity. Recently, accumulated evidences suggest that the inhibitory effect of GABA is determined by low intracellular chloride concentration, which is predominantly mediated by KCC2. This study was designed to investigate whether KCC2-GABAA pathway was involved in the mechanism underlying acupuncture intervention in rats with middle cerebral artery occlusion (MCAO). Three days after modeling, the rats received waggle needling, routine needling and placebo needling for 7 consecutive days. After treatment, the muscle spasticity, motor function and infarct volumes were tested. KCC2 and GABAAγ2 levels were detected via western blotting, RT-PCR and immunofluorescence. KCC2 antagonist and agonist were administered after the last intervention. We found that acupuncture, particularly waggle needling, could remarkably alleviate muscle spasticity, reverse motor deficits and reduce cerebral infraction in MCAO rats, possibly due to its effects on up-regulating expressions of KCC2 and GABAAγ2 in the cortical infarct regions. However, the effects were blocked by KCC2 antagonist. In summary, this study suggests that improvements in muscle spasticity and motor function induced by waggle needling correlates with the activation of KCC2-GABAA pathway.


Subject(s)
Acupuncture Therapy/methods , Infarction, Middle Cerebral Artery/therapy , Muscle, Skeletal/metabolism , Stroke Rehabilitation/methods , Acupuncture Points , Animals , Infarction, Middle Cerebral Artery/rehabilitation , Male , Muscle Spasticity/therapy , Muscle, Skeletal/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , Symporters/genetics , Symporters/metabolism , Up-Regulation , K Cl- Cotransporters
5.
Mol Psychiatry ; 26(2): 383-395, 2021 02.
Article in English | MEDLINE | ID: mdl-33432190

ABSTRACT

The GABAB receptor (GABABR) agonist baclofen has been used to treat alcohol and several other substance use disorders (AUD/SUD), yet its underlying neural mechanism remains unclear. The present study aimed to investigate cortical GABABR dynamics following chronic alcohol exposure. Ex vivo brain slice recordings from mice chronically exposed to alcohol revealed a reduction in GABABR-mediated currents, as well as a decrease of GABAB1/2R and G-protein-coupled inwardly rectifying potassium channel 2 (GIRK2) activities in the motor cortex. Moreover, our data indicated that these alterations could be attributed to dephosphorylation at the site of serine 783 (ser-783) in GABAB2 subunit, which regulates the surface expression of GABABR. Furthermore, a human study using paired-pulse-transcranial magnetic stimulation (TMS) analysis further demonstrated a reduced cortical inhibition mediated by GABABR in patients with AUD. Our findings provide the first evidence that chronic alcohol exposure is associated with significantly impaired cortical GABABR function. The ability to promote GABABR signaling may account for the therapeutic efficacy of baclofen in AUD.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels , Motor Cortex , Animals , Baclofen/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Humans , Mice , Receptors, GABA-B/metabolism , Signal Transduction
6.
Elife ; 92020 09 09.
Article in English | MEDLINE | ID: mdl-32902384

ABSTRACT

Absence seizures result from 3 to 5 Hz generalized thalamocortical oscillations that depend on highly regulated inhibitory neurotransmission in the thalamus. Efficient reuptake of the inhibitory neurotransmitter GABA is essential, and reuptake failure worsens human seizures. Here, we show that blocking GABA transporters (GATs) in acute rat brain slices containing key parts of the thalamocortical seizure network modulates epileptiform activity. As expected, we found that blocking either GAT1 or GAT3 prolonged oscillations. However, blocking both GATs unexpectedly suppressed oscillations. Integrating experimental observations into single-neuron and network-level computational models shows how a non-linear dependence of T-type calcium channel gating on GABAB receptor activity regulates network oscillations. Receptor activity that is either too brief or too protracted fails to sufficiently open T-type channels necessary for sustaining oscillations. Only within a narrow range does prolonging GABAB receptor activity promote channel opening and intensify oscillations. These results have implications for therapeutics that modulate inhibition kinetics.


Subject(s)
Calcium Channels, T-Type/metabolism , Models, Neurological , Neurons/physiology , Thalamus/physiology , Animals , Cells, Cultured , GABA Plasma Membrane Transport Proteins/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/metabolism , Seizures/metabolism
7.
Eur J Pharmacol ; 884: 173437, 2020 Oct 05.
Article in English | MEDLINE | ID: mdl-32735986

ABSTRACT

The GABA analog phenibut (ß-Phenyl-GABA) is a GABAB receptor agonist that has been licensed for various uses in Russia. Phenibut is also available as a dietary supplement from online vendors worldwide, and previous studies have indicated that phenibut overdose results in intoxication, withdrawal symptoms, and addiction. F-phenibut (ß-(4-Fluorophenyl)-GABA), a derivative of phenibut, has not been approved for clinical use. However, it is also available as a nootropic supplement from online suppliers. F-phenibut binds to GABAB with a higher affinity than phenibut; therefore, F-phenibut may lead to more serious intoxication than phenibut. However, the mechanisms by which F-phenibut acts on GABAB receptors and influences neuronal function remain unknown. In the present study, we compared the potency of F-phenibut, phenibut, and the GABAB agonist (±)-baclofen (baclofen) using in vitro patch-clamp recordings obtained from mouse cerebellar Purkinje cells slice preparations Our findings indicate that F-phenibut acted as a potent GABAB agonist. EC50 of outward current density evoked by the three GABAB agonists decreased in the following order: phenibut (1362 µM) > F-phenibut (23.3 µM) > baclofen (6.0 µM). The outward current induced by GABAB agonists was an outward-rectifying K+ current, in contrast to the previous finding that GABAB agonists activates an inward-rectifying K+ current. The K+ current recorded in the present study was insensitive to extracellular Ba2+, intra- or extracellular Cs+, and intra- or extracellular tetraethylammonium-Cl. Moreover, F-phenibut suppressed action potential generation in Purkinje cells. Thus, abuse of F-phenibut may lead to severe damage by inhibiting the excitability of GABAB-expressing neurons.


Subject(s)
GABA-B Receptor Agonists/pharmacology , Potassium Channels/metabolism , Potassium/metabolism , Purkinje Cells/drug effects , Receptors, GABA-B/drug effects , gamma-Aminobutyric Acid/pharmacology , Action Potentials , Animals , Baclofen/pharmacology , Dose-Response Relationship, Drug , Female , GABA-B Receptor Agonists/toxicity , In Vitro Techniques , Male , Mice, Inbred ICR , Purkinje Cells/metabolism , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , gamma-Aminobutyric Acid/toxicity
8.
Bioorg Med Chem Lett ; 30(18): 127443, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32730942

ABSTRACT

Positive allosteric modulators (PAMs) of GABAB receptor represent an interesting alternative to receptor agonists such as baclofen, as they act on the receptor in a more physiological way and thus are devoid of the side effects typically exerted by the agonists. Based on our interest in the identification of new GABAB receptor PAMs, we followed a merging approach to design new chemotypes starting from selected active compounds, such as GS39783, rac-BHFF, and BHF177, and we ended up with the synthesis of four different classes of compounds. The new compounds were tested alone or in the presence of 10 µM GABA using [35S]GTPγS binding assay to assess their functionality at the receptor. Unexpectedly, a number of them significantly inhibited GABA-stimulated GTPγS binding thus revealing a functional switch with respect to the prototype molecules. Further studies on selected compounds will clarify if they act as negative modulators of the receptor or, instead, as antagonists at the orthosteric binding site.


Subject(s)
Baclofen/chemical synthesis , GABA-B Receptor Agonists/chemical synthesis , Guanosine 5'-O-(3-Thiotriphosphate)/chemistry , Receptors, GABA-B/metabolism , Allosteric Regulation , Baclofen/metabolism , Benzofurans/pharmacology , Binding Sites , Cyclization , Cyclopentanes/pharmacology , Drug Evaluation, Preclinical , GABA Modulators/metabolism , GABA-B Receptor Agonists/metabolism , Humans , Norbornanes/pharmacology , Protein Binding , Pyrimidines/pharmacology , Structure-Activity Relationship
9.
Nature ; 584(7820): 304-309, 2020 08.
Article in English | MEDLINE | ID: mdl-32581365

ABSTRACT

The human GABAB receptor-a member of the class C family of G-protein-coupled receptors (GPCRs)-mediates inhibitory neurotransmission and has been implicated in epilepsy, pain and addiction1. A unique GPCR that is known to require heterodimerization for function2-6, the GABAB receptor has two subunits, GABAB1 and GABAB2, that are structurally homologous but perform distinct and complementary functions. GABAB1 recognizes orthosteric ligands7,8, while GABAB2 couples with G proteins9-14. Each subunit is characterized by an extracellular Venus flytrap (VFT) module, a descending peptide linker, a seven-helix transmembrane domain and a cytoplasmic tail15. Although the VFT heterodimer structure has been resolved16, the structure of the full-length receptor and its transmembrane signalling mechanism remain unknown. Here we present a near full-length structure of the GABAB receptor, captured in an inactive state by cryo-electron microscopy. Our structure reveals several ligands that preassociate with the receptor, including two large endogenous phospholipids that are embedded within the transmembrane domains to maintain receptor integrity and modulate receptor function. We also identify a previously unknown heterodimer interface between transmembrane helices 3 and 5 of both subunits, which serves as a signature of the inactive conformation. A unique 'intersubunit latch' within this transmembrane interface maintains the inactive state, and its disruption leads to constitutive receptor activity.


Subject(s)
Cryoelectron Microscopy , Receptors, GABA-B/chemistry , Receptors, GABA-B/ultrastructure , Calcium/metabolism , Ethanolamines/chemistry , Ethanolamines/metabolism , Humans , Ligands , Models, Molecular , Phosphorylcholine/chemistry , Phosphorylcholine/metabolism , Protein Domains , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, GABA-B/metabolism , Structure-Activity Relationship
10.
Brain Res Bull ; 149: 240-250, 2019 07.
Article in English | MEDLINE | ID: mdl-31034945

ABSTRACT

Orofacial inflammation could activate satellite glial cells (SGCs) in the trigeminal ganglion (TG) to produce interleukin 1ß (IL-1ß) which plays crucial roles in the development of inflammatory pain. Recent studies have shown that gamma-amino butyric acid-B (GABAB) receptor could modulate the expression of inflammatory cytokines in microglia and astrocytes in the spinal cord. The objective of this study was to investigate whether GABAB receptors in TG SGCs attenuate inflammatory facial pain via mediating IL-1ß following inflammation and its mechanisms. Complete Freund's adjuvant (CFA) was injected into the whisker pad of rats to induce inflammation in vivo. Lipopolysaccharide (LPS) was added to culture medium to activate SGCs in vitro. Behavioral measures showed that microinjection of baclofen (a selective GABAB receptor agonist) into the TG ameliorated the mechanical allodynia of CFA-treated rats. Interestingly, baclofen pretreatment inhibited SGC activation and IL-1ß production, however, preserved the decreased expression of GABAB receptors in SGCs activated by CFA in vivo and LPS in vitro. In addition, baclofen suppressed the increased expression of p-NF- κ B p65, p-I κ Bα, and p-p38 MAPK, while reversed the decreased production of I κ Bα, and further enhanced the increased expression of p-ERK(1/2) in LPS-treated SGCs in vitro. Finally, those effects of baclofen were abolished by saclofen (a specific GABAB receptor antagonist) co-administration. Altogether, these results demonstrated for the first time that activation of GABAB receptor might inhibit IL-1ß production by suppressing NF- κ B and p38 MAPK signaling pathway activation and restore GABAB receptor expression in SGCs to attenuate inflammatory facial pain.


Subject(s)
Facial Pain/metabolism , Interleukin-1beta/metabolism , Receptors, GABA-B/metabolism , Animals , Baclofen/pharmacology , Cytokines/metabolism , Facial Pain/physiopathology , GABA-B Receptor Agonists/pharmacology , Hyperalgesia/metabolism , Inflammation , MAP Kinase Signaling System/physiology , Male , Microglia/metabolism , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Neuroglia/metabolism , Neuroglia/physiology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Satellite Cells, Perineuronal/metabolism , Signal Transduction/physiology , Trigeminal Ganglion/metabolism , gamma-Aminobutyric Acid/metabolism
11.
Neurorehabil Neural Repair ; 33(2): 130-140, 2019 02.
Article in English | MEDLINE | ID: mdl-30744527

ABSTRACT

BACKGROUND: Stroke is a leading cause of adult disability owing largely to motor impairment and loss of function. After stroke, there may be abnormalities in γ-aminobutyric acid (GABA)-mediated inhibitory function within primary motor cortex (M1), which may have implications for residual motor impairment and the potential for functional improvements at the chronic stage. OBJECTIVE: To quantify GABA neurotransmission and concentration within ipsilesional and contralesional M1 and determine if they relate to upper limb impairment and function at the chronic stage of stroke. METHODS: Twelve chronic stroke patients and 16 age-similar controls were recruited for the study. Upper limb impairment and function were assessed with the Fugl-Meyer Upper Extremity Scale and Action Research Arm Test. Threshold tracking paired-pulse transcranial magnetic stimulation protocols were used to examine short- and long-interval intracortical inhibition and late cortical disinhibition. Magnetic resonance spectroscopy was used to evaluate GABA concentration. RESULTS: Short-interval intracortical inhibition was similar between patients and controls ( P = .10). Long-interval intracortical inhibition was greater in ipsilesional M1 compared with controls ( P < .001). Patients who did not exhibit late cortical disinhibition in ipsilesional M1 were those with greater upper limb impairment and worse function ( P = .002 and P = .017). GABA concentration was lower within ipsilesional ( P = .009) and contralesional ( P = .021) M1 compared with controls, resulting in an elevated excitation-inhibition ratio for patients. CONCLUSION: These findings indicate that ipsilesional and contralesional M1 GABAergic inhibition are altered in this small cohort of chronic stroke patients. Further study is warranted to determine how M1 inhibitory networks might be targeted to improve motor function.


Subject(s)
Motor Cortex/physiopathology , Movement Disorders/physiopathology , Neural Inhibition , Stroke/physiopathology , Upper Extremity/physiopathology , Aged , Aged, 80 and over , Chronic Disease , Electromyography , Evoked Potentials, Motor , Female , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Middle Aged , Motor Cortex/diagnostic imaging , Movement Disorders/diagnostic imaging , Movement Disorders/etiology , Multimodal Imaging , Receptors, GABA-B/metabolism , Stroke/diagnostic imaging , Transcranial Magnetic Stimulation , gamma-Aminobutyric Acid/metabolism
12.
J Biol Chem ; 294(7): 2375-2385, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30545943

ABSTRACT

GABAA receptors (GABAARs) are pentameric ligand-gated ion channels that mediate synaptic inhibition throughout the central nervous system. The α1ß2γ2 receptor is the major subtype in the brain; GABA binds at the ß2(+)α1(-) interface. The structure of the homomeric ß3 GABAAR, which is not activated by GABA, has been solved. Recently, four additional heteromeric structures were reported, highlighting key residues required for agonist binding. Here, we used a protein engineering method, taking advantage of knowledge of the key binding residues, to create a ß3(+)α1(-) heteromeric interface in the homomeric human ß3 GABAAR that enables GABA-mediated activation. Substitutions were made in the complementary side of the orthosteric binding site in loop D (Y87F and Q89R), loop E (G152T), and loop G (N66D and A70T). The Q89R and G152T combination enabled low-potency activation by GABA and potentiation by propofol but impaired direct activation by higher propofol concentrations. At higher concentrations, GABA inhibited gating of ß3 GABAAR variants containing Y87F, Q89R, and G152T. Reversion of Phe87 to tyrosine abolished GABA's inhibitory effect and partially recovered direct activation by propofol. This tyrosine is conserved in homomeric GABAARs and in the Erwinia chrysanthemi ligand-gated ion channel and may be essential for the absence of an inhibitory effect of GABA on homomeric channels. This work demonstrated that only two substitutions, Q89R and G152T, in ß3 GABAAR are sufficient to reconstitute GABA-mediated activation and suggests that Tyr87 prevents inhibitory effects of GABA.


Subject(s)
Ion Channel Gating , Mutation, Missense , Protein Structure, Secondary , Receptors, GABA-B , Amino Acid Substitution , Catalytic Domain , Dickeya chrysanthemi/chemistry , Dickeya chrysanthemi/genetics , Dickeya chrysanthemi/metabolism , HEK293 Cells , Humans , Propofol/pharmacology , Receptors, GABA-B/chemistry , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/metabolism
13.
BMC Cancer ; 18(1): 263, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29514603

ABSTRACT

BACKGROUND: High-grade chondrosarcoma, which has a high incidence of local recurrence and pulmonary metastasis despite surgical resection, is associated with poor prognosis. Therefore, new and effective adjuvant therapies are urgently required for this disease. Gamma-aminobutyric acid (GABA), which acts as a neurotrophic factor during nervous system development, is related to the proliferation and migration of certain cancer cells. The GABAergic system, which is composed of GABA, the GABA-synthesizing enzyme glutamic acid decarboxylase (GAD), and GABA receptors, has an important function in nerve growth and development of neural crest. Therefore, the GABAergic system may play important functional roles in the proliferation of chondrosarcoma cells, which are derived from neural crest cells. We examined the anti-tumor effects of the GABAergic system on a chondrosarcoma cell line. METHODS: We evaluated the underlying mechanisms of the anti-tumor effects of the GABAergic system, such as the involvement of different signaling pathways, apoptosis, and cell cycle arrest, in the high-grade chondrosarcoma cell line OUMS-27. In addition, we performed whole-cell patch-clamp recordings for Ca2+ currents and evaluated the changes in intracellular Ca2+ concentration via Ca2+ channels, which are related to the GABAB receptor in high-grade chondrosarcoma cells. RESULTS: The GABAB receptor antagonist CGP had anti-tumor effects on high-grade chondrosarcoma cells in a dose-dependent manner. The activities of caspase 3 and caspase 9 were significantly elevated in CGP-treated cells compared to in untreated cells. The activity of caspase 8 did not differ significantly between untreated cells and CGP-treated cells. However, caspase 8 tended to be up-regulated in CGP-treated cells. The GABAB receptor antagonist exhibited anti-tumor effects at the G1/S cell cycle checkpoint and induced apoptosis via dual inhibition of the PI3/Akt/mTOR and MAPK signaling pathways. Furthermore, the changes in intracellular Ca2+ via GABAB receptor-related Ca2+ channels inhibited the proliferation of high-grade chondrosarcoma cells by inducing and modulating apoptotic pathways. CONCLUSIONS: The GABAB receptor antagonist may improve the prognosis of high-grade chondrosarcoma by exerting anti-tumor effects via different signaling pathways, apoptosis, cell cycle arrest, and Ca2+ channels in high-grade chondrosarcoma cells.


Subject(s)
Apoptosis , Bone Neoplasms/pathology , Calcium/metabolism , Cell Proliferation , Chondrosarcoma/pathology , Receptors, GABA-B/metabolism , Bone Neoplasms/metabolism , Cell Cycle , Chondrosarcoma/metabolism , GABA-B Receptor Antagonists/pharmacology , Humans , Patch-Clamp Techniques , Receptors, GABA-B/chemistry , Signal Transduction , Tumor Cells, Cultured
14.
Neuropharmacology ; 136(Pt A): 106-116, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29106983

ABSTRACT

Following the discovery of GABAB receptors by Norman Bowery and colleagues, cloning and biochemical efforts revealed that GABAB receptors assemble multi-subunit complexes composed of principal and auxiliary subunits. The principal receptor subunits GABAB1a, GABAB1b and GABAB2 form two heterodimeric GABAB(1a,2) and GABAB(1b,2) receptors that can associate with tetramers of auxiliary KCTD (K+ channel tetramerization domain) subunits. Experiments with subunit knock-out mice revealed that GABAB(1b,2) receptors activate slow inhibitory postsynaptic currents (sIPSCs) while GABAB(1a,2) receptors function as heteroreceptors and inhibit glutamate release. Both GABAB(1a,2) and GABAB(1b,2) receptors can serve as autoreceptors and inhibit GABA release. Auxiliary KCTD subunits regulate the duration of sIPSCs and scaffold effector channels at the receptor. GABAB receptors are well known to contribute to thalamic spindle oscillations. Spindles are generated through alternating burst-firing in reciprocally connected glutamatergic thalamocortical relay (TCR) and GABAergic thalamic reticular nucleus (TRN) neurons. The available data implicate postsynaptic GABAB receptors in TCR cells in the regulation of spindle frequency. We now used electrical or optogenetic activation of thalamic spindles and pharmacological experiments in acute slices of knock-out mice to study the impact of GABAB(1a,2) and GABAB(1b,2) receptors on spindle oscillations. We found that selectively GABAB(1a,2) heteroreceptors at TCR to TRN cell synapses regulate oscillation strength, while GABAB(1b,2) receptors control oscillation frequency. The auxiliary subunit KCTD16 influences both oscillation strength and frequency, supporting that KCTD16 regulates network activity through GABAB(1a,2) and GABAB(1b,2) receptors. This article is part of the "Special Issue Dedicated to Norman G. Bowery".


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, GABA-B/metabolism , Thalamus/metabolism , Animals , Glutamic Acid/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurons/metabolism , Receptors, GABA/deficiency , Receptors, GABA/genetics , Receptors, GABA-B/genetics , Synapses/metabolism , Tissue Culture Techniques
15.
Drug Metab Dispos ; 46(1): 53-65, 2018 01.
Article in English | MEDLINE | ID: mdl-29061584

ABSTRACT

A bidirectional route of communication between the gastrointestinal tract and the central nervous system, termed the "gut-brain axis," is becoming increasingly relevant to treatment of cerebral damage. Panax Notoginsenoside extract (PNE) is popular for prevention and treatment of cardio-cerebrovascular ischemic diseases although plasma and cerebral exposure levels are extremely low. To date, the mechanisms underlying the neuroprotective effects of PNE remain largely unknown. In the present study, the neuroprotective effects of PNE were systematically studied via investigation of the regulation by PNE of the gastrointestinal microbial community and γ aminobutyric acid (GABA) receptors. The results demonstrated that pretreatment with PNE exerted a remarkable neuroprotective effect on focal cerebral ischemia/reperfusion (I/R) injury in rats, and the efficiency was attenuated in germ-free rats. Pretreatment with PNE could significantly prevent downregulation of Bifidobacterium longum (B.L) caused by I/R surgery, and colonization by B.L could also exert neuroprotective effects. More importantly, both PNE and B.L could upregulate the expression of GABA receptors in the hippocampus of I/R rats, and coadministration of a GABA-B receptor antagonist could significantly attenuate the neuroprotective effects of PNE and B.L. The study above suggests that the neuroprotective effects of PNE may be largely attributable to its regulation of intestinal flora, and oral treatment with B.L was also useful in therapy of ischemia/reperfusion injury (I/R) by upregulating GABA-B receptors.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Hypoxia-Ischemia, Brain/prevention & control , Neuroprotective Agents/pharmacology , Panax/chemistry , Reperfusion Injury/prevention & control , Animals , Bifidobacterium longum/drug effects , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , GABA-B Receptor Antagonists/pharmacology , Gastrointestinal Microbiome/physiology , Ginsenosides/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hypoxia-Ischemia, Brain/etiology , Intestines/drug effects , Intestines/microbiology , Intestines/physiology , Neuroprotective Agents/chemistry , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/metabolism , Reperfusion Injury/etiology , Tissue Distribution , Up-Regulation
16.
Channels (Austin) ; 11(6): 574-586, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28742985

ABSTRACT

Burst-firing in thalamic neurons is known to play a key role in mediating thalamocortical (TC) oscillations that are associated with non-REM sleep and some types of epileptic seizure. Within the TC system the primary output of GABAergic neurons in the reticular thalamic nucleus (RTN) is thought to induce the de-inactivation of T-type calcium channels in thalamic relay (TR) neurons, promoting burst-firing drive to the cortex and the propagation of TC network activity. However, RTN neurons also project back onto other neurons within the RTN. The role of this putative negative feedback upon the RTN itself is less well understood, although is hypothesized to induce de-synchronization of RTN neuron firing leading to the suppression of TC oscillations. Here we tested two hypotheses concerning possible mechanisms underlying TC oscillation modulation. Firstly, we assessed the burst-firing behavior of RTN neurons in response to GABAB receptor activation using acute brain slices. The selective GABAB receptor agonist baclofen was found to induce suppression of burst-firing concurrent with effects on membrane input resistance. Secondly, RTN neurons express CaV3.2 and CaV3.3 T-type calcium channel isoforms known to contribute toward TC burst-firing and we examined the modulation of these channels by GABAB receptor activation. Utilizing exogenously expressed T-type channels we assessed whether GABAB receptor activation could directly alter T-type calcium channel properties. Overall, GABAB receptor activation had only modest effects on CaV3.2 and CaV3.3 isoforms. The only effect that could be predicted to suppress burst-firing was a hyperpolarized shift in the voltage-dependence of inactivation, potentially causing lower channel availability at membrane potentials critical for burst-firing. Conversely, other effects observed such as a hyperpolarized shift in the voltage-dependence of activation of both CaV3.2 and CaV3.3 as well as increased time constant of activation of the CaV3.3 isoform would be expected to enhance burst-firing. Together, we hypothesize that GABAB receptor activation mediates multiple downstream effectors that combined act to suppress burst-firing within the RTN. It appears unlikely that direct GABAB receptor-mediated modulation of T-type calcium channels is the major mechanistic contributor to this suppression.


Subject(s)
Neurons/metabolism , Receptors, GABA-B/metabolism , Thalamus/cytology , Animals , Calcium Channels, T-Type/metabolism , Female , Male , Rats , Rats, Wistar
17.
Am J Physiol Regul Integr Comp Physiol ; 312(3): R292-R300, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27974317

ABSTRACT

This study investigated the role of γ-aminobutyric acid subtype B (GABAB) receptors in tibial and pudendal neuromodulation of bladder overactivity induced by intravesical administration of dilute (0.5%) acetic acid (AA) in α-chloralose-anesthetized cats. To inhibit bladder overactivity, tibial or pudendal nerve stimulation (TNS or PNS) was applied at 5 Hz and two or four times threshold (T) intensity for inducing toe or anal sphincter twitch. TNS at 2T or 4T intensity significantly (P < 0.05) increased the bladder capacity to 173.8 ± 16.2 or 198.5 ± 24.1%, respectively, of control capacity. Meanwhile, PNS at 2T or 4T intensity significantly (P < 0.05) increased the bladder capacity to 217 ± 18.8 and 221.3 ± 22.3% of control capacity, respectively. CGP52432 (a GABAB receptor antagonist) at intravenous dosages of 0.1-1 mg/kg completely removed the TNS inhibition in female cats but had no effect in male cats. CGP52432 administered intravenously also had no effect on control bladder capacity or the pudendal inhibition of bladder overactivity. These results reveal a sex difference in the role of GABAB receptors in tibial neuromodulation of bladder overactivity in cats and that GABAB receptors are not involved in either pudendal neuromodulation or irritation-induced bladder overactivity.


Subject(s)
Electric Stimulation Therapy/methods , Receptors, GABA-B/metabolism , Tibial Nerve/physiopathology , Urinary Bladder, Overactive/prevention & control , Urinary Bladder, Overactive/physiopathology , Urinary Bladder/physiopathology , Animals , Cats , Female , Male , Pudendal Nerve/physiology , Receptors, Neurotransmitter/metabolism , Sex Characteristics , Treatment Outcome , Urinary Bladder/innervation
18.
Neurosci Lett ; 621: 62-67, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27080427

ABSTRACT

Recent studies demonstrated that treatment with saikosaponin A (SSA) - an active ingredient of the medicinal herb, Bupleurum falcatum L. - selectively suppressed, likely via a GABAB receptor-mediated mechanism, intravenous self-administration of morphine and cocaine in rats [Yoon et al., 2012; 2013]. The present study was designed to investigate whether the capacity of SSA to suppress morphine and cocaine self-administration extends to oral alcohol self-administration. To this end, selectively bred Sardinian alcohol-preferring (sP) rats were trained to lever-respond on a Fixed Ratio (FR) 4 (FR4) schedule of reinforcement for alcohol (15%, v/v) in daily 30-min sessions. Once responding had stabilized, rats were tested under the FR4 (measure of alcohol reinforcing properties) and Progressive Ratio (PR; measure of alcohol motivational properties) schedules of reinforcement. The possible involvement of the GABAB receptor system was investigated testing the effect of (a) pretreatment with the GABAB receptor antagonist, SCH50911, and (b) combined treatment with the positive allosteric modulator of the GABAB receptor, GS39783. Treatment with SSA (0, 0.25, 0.5, and 1mg/kg, i.p.) markedly reduced lever-responding for alcohol, amount of self-administered alcohol, and breakpoint for alcohol (defined as the lowest response requirement not achieved in the PR experiment). Pretreatment with 2mg/kg SCH50911 (i.p.) resulted in a partial blockade of the reducing effect of 0.5mg/kg SSA on lever-responding for alcohol and amount of self-administered alcohol. Combination of per se ineffective doses of GS39783 (5mg/kg, i.g.) and SSA (0.1mg/kg, i.p.) reduced lever-responding for alcohol and amount of self-administered alcohol. These results (a) extend to alcohol self-administration the capacity of SSA to suppress morphine and cocaine self-administration in rats and (b) suggest that the GABAB receptor system is likely part of the neural substrate underlying the reducing effect of SSA on alcohol self-administration.


Subject(s)
Bupleurum/chemistry , Ethanol/pharmacology , Oleanolic Acid/analogs & derivatives , Receptors, GABA-B/metabolism , Saponins/pharmacology , Allosteric Regulation , Animals , Cyclopentanes/pharmacology , Ethanol/administration & dosage , GABA-B Receptor Antagonists/pharmacology , Male , Morpholines/pharmacology , Motivation , Motor Activity/drug effects , Oleanolic Acid/pharmacology , Pyrimidines/pharmacology , Rats , Reinforcement, Psychology , Self Administration
19.
Brain Res ; 1647: 94-104, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27125595

ABSTRACT

Healthy neurons have an optimal operating range, coded globally by the frequency of action potentials or locally by calcium. The maintenance of this range is governed by homeostatic plasticity. Here, we discuss how new approaches to treat depression alter synaptic activity. These approaches induce the neuron to recruit homeostatic mechanisms to relieve depression. Homeostasis generally implies that the direction of activity necessary to restore the neuron's critical operating range is opposite in direction to its current activity pattern. Unconventional antidepressant therapies-deep brain stimulation and NMDAR antagonists-alter the neuron's "depressed" state by pushing the neuron's current activity in the same direction but to the extreme edge. These therapies rally the intrinsic drive of neurons in the opposite direction, thereby allowing the cell to return to baseline activity, form new synapses, and restore proper communication. In this review, we discuss seminal studies on protein synthesis dependent homeostatic plasticity and their contribution to our understanding of molecular mechanisms underlying the effectiveness of NMDAR antagonists as rapid antidepressants. Rapid antidepressant efficacy is likely to require a cascade of mRNA translational regulation. Emerging evidence suggests that changes in synaptic strength or intrinsic excitability converge on the same protein synthesis pathways, relieving depressive symptoms. Thus, we address the question: Are there multiple homeostatic mechanisms that induce the neuron and neuronal circuits to self-correct to regulate mood in vivo? Targeting alternative ways to induce homeostatic protein synthesis may provide, faster, safer, and longer lasting antidepressants. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.


Subject(s)
Antidepressive Agents/therapeutic use , Brain/drug effects , Depressive Disorder/drug therapy , Homeostasis/drug effects , Neurons/drug effects , Protein Biosynthesis/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Antidepressive Agents/administration & dosage , Autophagy/drug effects , Brain/physiology , Depressive Disorder/metabolism , Humans , Neuronal Plasticity/drug effects , Neurons/physiology , Receptors, GABA-B/metabolism , TOR Serine-Threonine Kinases/metabolism
20.
J Neurophysiol ; 115(5): 2376-88, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26912590

ABSTRACT

Whereas the activation of Gαi/o-coupled receptors commonly results in postsynaptic responses that show acute desensitization, the presynaptic inhibition of transmitter release caused by many Gαi/o-coupled receptors is maintained during agonist exposure. However, an exception has been noted where GABAB receptor (GABABR)-mediated inhibition of inhibitory postsynaptic currents (IPSCs) recorded in mouse proopiomelanocortin (POMC) neurons exhibit acute desensitization in ∼25% of experiments. To determine whether differential effector coupling confers sensitivity to desensitization, voltage-clamp recordings were made from POMC neurons to compare the mechanism by which µ-opioid receptors (MORs) and GABABRs inhibit transmitter release. Neither MOR- nor GABABR-mediated inhibition of release relied on the activation of presynaptic K(+) channels. Both receptors maintained the ability to inhibit release in the absence of external Ca(2+) or in the presence of ionomycin-induced Ca(2+) influx, indicating that inhibition of release can occur through a Ca(2+)-independent mechanism. Replacing Ca(2+) with Sr(2+) to disrupt G-protein-mediated inhibition of release occurring directly at the release machinery did not alter MOR- or GABAB -mediated inhibition of IPSCs, suggesting that reductions in evoked release can occur through the inhibition of Ca(2+) channels. Additionally, both receptors inhibited evoked IPSCs in the presence of selective blockers of N- or P/Q-type Ca(2+) channels. Altogether, the results show that MORs and GABABRs can inhibit transmitter release through the inhibition of calcium influx and by direct actions at the release machinery. Furthermore, since both the desensitizing and nondesensitizing presynaptic receptors are similarly coupled, differential effector coupling is unlikely responsible for differential desensitization of the inhibition of release.


Subject(s)
Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hypothalamus/physiology , Inhibitory Postsynaptic Potentials , Receptors, GABA-B/metabolism , Synapses/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Calcium Channels/metabolism , Calcium Signaling , Mice , Mice, Inbred C57BL , Receptors, Opioid, mu/metabolism , Synapses/drug effects , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL