Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 111(29): 10755-60, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002517

ABSTRACT

ML297 was recently identified as a potent and selective small molecule agonist of G-protein-gated inwardly rectifying K(+) (GIRK/Kir3) channels. Here, we show ML297 selectively activates recombinant neuronal GIRK channels containing the GIRK1 subunit in a manner that requires phosphatidylinositol-4,5-bisphosphate (PIP2), but is otherwise distinct from receptor-induced, G-protein-dependent channel activation. Two amino acids unique to the pore helix (F137) and second membrane-spanning (D173) domain of GIRK1 were identified as necessary and sufficient for the selective activation of GIRK channels by ML297. Further investigation into the behavioral effects of ML297 revealed that in addition to its known antiseizure efficacy, ML297 decreases anxiety-related behavior without sedative or addictive liabilities. Importantly, the anxiolytic effect of ML297 was lost in mice lacking GIRK1. Thus, activation of GIRK1-containing channels by ML297 or derivatives may represent a new approach to the treatment of seizure and/or anxiety disorders.


Subject(s)
Anti-Anxiety Agents/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ion Channel Gating/drug effects , Phenylurea Compounds/pharmacology , Pyrazoles/pharmacology , Amino Acid Sequence , Animals , Baclofen/pharmacology , Behavior, Animal/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neurons/drug effects , Neurons/metabolism
2.
J Neurochem ; 130(5): 642-56, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24666322

ABSTRACT

HIV-associated neurocognitive disorders afflict about half of HIV-infected patients. HIV-infected cells shed viral proteins, such as the transactivator of transcription (Tat), which can cause neurotoxicity by over activation of NMDA receptors. Here, we show that Tat causes a time-dependent, biphasic change in NMDA-evoked increases in intracellular Ca(2+) concentration ([Ca(2+)]i). NMDA-evoked responses were potentiated following 2-h exposure to Tat (50 ng/mL). Tat-induced potentiation of NMDA-evoked increases in [Ca(2+)]i peaked by 8 h and then adapted by gradually reversing to baseline by 24 h and eventually dropping below control by 48 h. Tat-induced potentiation of NMDA-evoked responses was blocked by inhibition of lipoprotein receptor-related protein (LRP) or Src tyrosine kinase. Potentiation was unaffected by inhibition of nitric oxide synthase (NOS). However, NOS activity was required for adaptation. Adaptation was also prevented by inhibition of soluble guanylate cyclase (sGC) and cyclic guanosine monophosphate-dependent protein kinase G (PKG). Together, these findings indicate that Tat potentiates NMDA-evoked increases in [Ca(2+)]i via LRP-dependent activation of Src and that this potentiation adapts via activation of the NOS/sGC/PKG pathway. Adaptation may protect neurons from excessive Ca(2+) influx and could reveal targets for the treatment of HIV-associated neurocognitive disorders. HIV-associated neurocognitive disorders (HAND) afflict about half of HIV-infected patients. HIV-infected cells shed viral proteins, such as the transactivator of transcription (Tat), which can cause neurotoxicity by over activation of NMDA receptors (NMDARs). We show that HIV-1 Tat evoked biphasic changes in NMDA-evoked [Ca(2+) ]i responses. Initially, Tat potentiated NMDA-evoked responses following LRP-mediated activation of Src kinase. Subsequently, Tat-induced NMDAR potentiation adapted by activation of a NOS/sGC/PKG pathway that attenuated NMDA-evoked increases in [Ca(2+)]i . Adaptation may be a novel neuroprotective mechanism to prevent excessive Ca(2+) influx. Solid and dashed arrows represent direct and potentially indirect connections, respectively.


Subject(s)
Calcium/metabolism , Nitric Oxide/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/physiology , src-Family Kinases/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Cells, Cultured , HIV Infections/metabolism , HIV-1/metabolism , Immunoblotting , N-Methylaspartate/metabolism , Neurons/metabolism , Neurons/virology , Rats , Transfection
3.
J Biol Chem ; 289(4): 2440-9, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24318880

ABSTRACT

Parasympathetic activity decreases heart rate (HR) by inhibiting pacemaker cells in the sinoatrial node (SAN). Dysregulation of parasympathetic influence has been linked to sinus node dysfunction and arrhythmia. RGS (regulator of G protein signaling) proteins are negative modulators of the parasympathetic regulation of HR and the prototypical M2 muscarinic receptor (M2R)-dependent signaling pathway in the SAN that involves the muscarinic-gated atrial K(+) channel IKACh. Both RGS4 and RGS6-Gß5 have been implicated in these processes. Here, we used Rgs4(-/-), Rgs6(-/-), and Rgs4(-/-):Rgs6(-/-) mice to compare the relative influence of RGS4 and RGS6 on parasympathetic regulation of HR and M2R-IKACh-dependent signaling in the SAN. In retrogradely perfused hearts, ablation of RGS6, but not RGS4, correlated with decreased resting HR, increased heart rate variability, and enhanced sensitivity to the negative chronotropic effects of the muscarinic agonist carbachol. Similarly, loss of RGS6, but not RGS4, correlated with enhanced sensitivity of the M2R-IKACh signaling pathway in SAN cells to carbachol and a significant slowing of M2R-IKACh deactivation rate. Surprisingly, concurrent genetic ablation of RGS4 partially rescued some deficits observed in Rgs6(-/-) mice. These findings, together with those from an acute pharmacologic approach in SAN cells from Rgs6(-/-) and Gß5(-/-) mice, suggest that the partial rescue of phenotypes in Rgs4(-/-):Rgs6(-/-) mice is attributable to another R7 RGS protein whose influence on M2R-IKACh signaling is masked by RGS4. Thus, RGS6-Gß5, but not RGS4, is the primary RGS modulator of parasympathetic HR regulation and SAN M2R-IKACh signaling in mice.


Subject(s)
Heart Rate/physiology , Muscle Proteins/metabolism , Parasympathetic Nervous System/metabolism , RGS Proteins/metabolism , Sinoatrial Node/metabolism , Animals , Carbachol/pharmacology , Cardiotonic Agents/pharmacology , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/metabolism , Heart Rate/drug effects , Mice , Mice, Knockout , Muscle Proteins/genetics , Potassium Channels/genetics , Potassium Channels/metabolism , RGS Proteins/genetics , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M2/metabolism , Sinoatrial Node/cytology
4.
Hippocampus ; 23(12): 1231-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23804514

ABSTRACT

In the hippocampus, signaling through G protein-coupled receptors is modulated by Regulators of G protein signaling (Rgs) proteins, which act to stimulate the rate of GTP hydrolysis, and consequently, G protein inactivation. The R7-Rgs subfamily selectively deactivates the G(i/o)-class of Gα subunits that mediate the action of several GPCRs. Here, we used co-immunoprecipitation, electrophysiology and immunoelectron microscopy techniques to investigate the formation of macromolecular complexes and spatial relationship of Rgs7/Gß5 complexes and its prototypical signaling partners, the GABAB receptor and Girk channel. Co-expression of recombinant GABAB receptors and Girk channels in combination with co-immunoprecipitation experiments established that the Rgs7/Gß5 forms complexes with GABAB receptors or Girk channels. Using electrophysiological experiments, we found that GABAB -Girk current deactivation kinetics was markedly faster in cells coexpressing Rgs7/Gß5. At the electron microscopic level, immunolabeling for Rgs7 and Gß5 proteins was found primarily in the dendritic layers of the hippocampus and showed similar distribution patterns. Immunoreactivity was mostly localized along the extrasynaptic plasma membrane of dendritic shafts and spines of pyramidal cells and, to a lesser extent, to that of presynaptic terminals. Quantitative analysis of immunogold particles for Rgs7 and Gß5 revealed an enrichment of the two proteins around excitatory synapses on dendritic spines, virtually identical to that of Girk2 and GABAB1 . These data support the existence of macromolecular complexes composed of GABAB receptor-G protein-Rgs7-Girk channels in which Rgs7 and Gß5 proteins may preferentialy modulate GABAB receptor signaling through the deactivation of Girk channels on dendritic spines. In contrast, Rgs7 and Girk2 were associated but mainly segregated from GABAB1 in dendritic shafts, where Rgs7/Gß5 signaling complexes might modulate Girk-dependent signaling via a different metabotropic receptor(s).


Subject(s)
CA1 Region, Hippocampal/cytology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein beta Subunits/metabolism , Neurons/metabolism , RGS Proteins/metabolism , Receptors, GABA/metabolism , Animals , Biophysics , Cell Line, Transformed , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Electric Stimulation , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , GTP-Binding Protein beta Subunits/genetics , GTP-Binding Protein beta Subunits/ultrastructure , Humans , Imaging, Three-Dimensional , Immunoprecipitation , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Microscopy, Immunoelectron , Neurons/ultrastructure , Patch-Clamp Techniques , RGS Proteins/genetics , RGS Proteins/ultrastructure , Receptors, GABA/genetics , Receptors, GABA/ultrastructure , Transfection
5.
Proc Natl Acad Sci U S A ; 109(52): 21492-7, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236146

ABSTRACT

G protein-gated inwardly rectifying K(+) (Girk/K(IR)3) channels mediate the inhibitory effect of many neurotransmitters on excitable cells. Girk channels are tetramers consisting of various combinations of four mammalian Girk subunits (Girk1 to -4). Although Girk1 is unable to form functional homomeric channels, its presence in cardiac and neuronal channel complexes correlates with robust channel activity. This study sought to better understand the potentiating influence of Girk1, using the GABA(B) receptor and Girk1/Girk2 heteromer as a model system. Girk1 did not increase the protein levels or alter the trafficking of Girk2-containing channels to the cell surface in transfected cells or hippocampal neurons, indicating that its potentiating influence involves enhancement of channel activity. Structural elements in both the distal carboxyl-terminal domain and channel core were identified as key determinants of robust channel activity. In the distal carboxyl-terminal domain, residue Q404 was identified as a key determinant of receptor-induced channel activity. In the Girk1 core, three unique residues in the pore (P) loop (F137, A142, Y150) were identified as a collective potentiating influence on both receptor-dependent and receptor-independent channel activity, exerting their influence, at least in part, by enhancing mean open time and single-channel conductance. Interestingly, the potentiating influence of the Girk1 P-loop is tempered by residue F162 in the second membrane-spanning domain. Thus, discontinuous and sometime opposing elements in Girk1 underlie the Girk1-dependent potentiation of receptor-dependent and receptor-independent heteromeric channel activity.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ion Channel Gating , Protein Subunits/chemistry , Protein Subunits/metabolism , Amino Acid Sequence , Animals , Baclofen/pharmacology , Glutamine/metabolism , HEK293 Cells , Humans , Ion Channel Gating/drug effects , Mice , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Receptors, GABA-B/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
6.
Circ Res ; 107(11): 1350-4, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20884879

ABSTRACT

RATIONALE: The parasympathetic reduction in heart rate involves the sequential activation of m2 muscarinic cholinergic receptors (m(2)Rs), pertussis toxin-sensitive (Gi/o) heterotrimeric G proteins, and the atrial potassium channel I(KACh). Molecular mechanisms regulating this critical signal transduction pathway are not fully understood. OBJECTIVE: To determine whether the G protein signaling regulator Rgs6/Gß5 modulates m(2)R-I(KACh) signaling and cardiac physiology. METHODS AND RESULTS: Cardiac expression of Rgs6, and its interaction with Gß5, was demonstrated by immunoblotting and immunoprecipitation. Rgs6(-/-) mice were generated by gene targeting, and the cardiac effects of Rgs6 ablation were analyzed by whole-cell recordings in isolated cardiomyocytes and ECG telemetry. Loss of Rgs6 yielded profound delays in m(2)R-I(KACh) deactivation kinetics in both neonatal atrial myocytes and adult sinoatrial nodal cells. Rgs6(-/-) mice exhibited mild resting bradycardia and altered heart rate responses to pharmacological manipulations that were consistent with enhanced m(2)R-I(KACh) signaling. CONCLUSIONS: The cardiac Rgs6/Gß5 complex modulates the timing of parasympathetic influence on atrial myocytes and heart rate in mice.


Subject(s)
GTP-Binding Protein beta Subunits/physiology , Heart Rate/physiology , Ion Channel Gating/physiology , Myocytes, Cardiac/physiology , Parasympathetic Fibers, Postganglionic/physiology , Potassium Channels, Voltage-Gated/physiology , RGS Proteins/physiology , Up-Regulation/physiology , Animals , Down-Regulation/genetics , Down-Regulation/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , HEK293 Cells , Heart Atria/cytology , Heart Atria/physiopathology , Heart Rate/genetics , Humans , Ion Channel Gating/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Signal Transduction/genetics , Signal Transduction/physiology , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL