Presynaptic localization of G protein isoforms in the efferent nerve terminals of the mammalian cochlea

1998 ◽  
Vol 116 (1-2) ◽  
pp. 1-9 ◽  
Author(s):  
Mauricio Kurc ◽  
Valerie Dodane ◽  
Decio S Pinto ◽  
Bechara Kachar
2020 ◽  
Vol 117 (25) ◽  
pp. 14522-14531
Author(s):  
Allison Anderson ◽  
Ikuo Masuho ◽  
Ezequiel Marron Fernandez de Velasco ◽  
Atsushi Nakano ◽  
Lutz Birnbaumer ◽  
...  

How G protein-coupled receptors (GPCRs) evoke specific biological outcomes while utilizing a limited array of G proteins and effectors is poorly understood, particularly in native cell systems. Here, we examined signaling evoked by muscarinic (M2R) and adenosine (A1R) receptor activation in the mouse sinoatrial node (SAN), the cardiac pacemaker. M2R and A1R activate a shared pool of cardiac G protein-gated inwardly rectifying K+(GIRK) channels in SAN cells from adult mice, but A1R-GIRK responses are smaller and slower than M2R-GIRK responses. Recordings from mice lacking Regulator of G protein Signaling 6 (RGS6) revealed that RGS6 exerts a GPCR-dependent influence on GIRK-dependent signaling in SAN cells, suppressing M2R-GIRK coupling efficiency and kinetics and A1R-GIRK signaling amplitude. Fast kinetic bioluminescence resonance energy transfer assays in transfected HEK cells showed that RGS6 prefers Gαoover Gαias a substrate for its catalytic activity and that M2R signals preferentially via Gαo, while A1R does not discriminate between inhibitory G protein isoforms. The impact of atrial/SAN-selective ablation of Gαoor Gαi2was consistent with these findings. Gαi2ablation had minimal impact on M2R-GIRK and A1R-GIRK signaling in SAN cells. In contrast, Gαoablation decreased the amplitude and slowed the kinetics of M2R-GIRK responses, while enhancing the sensitivity and prolonging the deactivation rate of A1R-GIRK signaling. Collectively, our data show that differences in GPCR-G protein coupling preferences, and the Gαosubstrate preference of RGS6, shape A1R- and M2R-GIRK signaling dynamics in mouse SAN cells.


2019 ◽  
Vol 10 (5) ◽  
pp. 2720-2728 ◽  
Author(s):  
Ting Yang Hsieh ◽  
Yi Chang ◽  
Su Jane Wang

Piperine is the crucial alkaloid component of black pepper (Piper nigrum Linn.) and has neuroprotective effects.


1998 ◽  
Vol 18 (9) ◽  
pp. 3282-3296 ◽  
Author(s):  
Anne K. Hennig ◽  
Douglas A. Cotanche

1998 ◽  
Vol 273 (51) ◽  
pp. 34580-34585 ◽  
Author(s):  
Hideto Oishi ◽  
Takuya Sasaki ◽  
Fumiko Nagano ◽  
Wataru Ikeda ◽  
Takeshi Ohya ◽  
...  

Author(s):  
Jane Dingus ◽  
William E. McIntire ◽  
Michael D. Wilcox ◽  
John D. Hildebrandt

2018 ◽  
Vol 314 (5) ◽  
pp. C616-C626 ◽  
Author(s):  
Muriel Nobles ◽  
David Montaigne ◽  
Sonia Sebastian ◽  
Lutz Birnbaumer ◽  
Andrew Tinker

G protein-gated inwardly rectifying K+ (GIRK) channels are the major inwardly rectifying K+ currents in cardiac atrial myocytes and an important determinant of atrial electrophysiology. Inhibitory G protein α-subunits can both mediate activation via acetylcholine but can also suppress basal currents in the absence of agonist. We studied this phenomenon using whole cell patch clamping in murine atria from mice with global genetic deletion of Gαi2, combined deletion of Gαi1/Gαi3, and littermate controls. We found that mice with deletion of Gαi2 had increased basal and agonist-activated currents, particularly in the right atria while in contrast those with Gαi1/Gαi3 deletion had reduced currents. Mice with global genetic deletion of Gαi2 had decreased action potential duration. Tissue preparations of the left atria studied with a multielectrode array from Gαi2 knockout mice showed a shorter effective refractory period, with no change in conduction velocity, than littermate controls. Transcriptional studies revealed increased expression of GIRK channel subunit genes in Gαi2 knockout mice. Thus different G protein isoforms have differential effects on GIRK channel behavior and paradoxically Gαi2 act to increase basal and agonist-activated GIRK currents. Deletion of Gαi2 is potentially proarrhythmic in the atria.


1999 ◽  
Vol 261 (2) ◽  
pp. 517-523 ◽  
Author(s):  
Gerald Gaibelet ◽  
Eliane Meilhoc ◽  
Joelle Riond ◽  
Isabelle Saves ◽  
Torsten Exner ◽  
...  

Physiology ◽  
2007 ◽  
Vol 22 (3) ◽  
pp. 174-184 ◽  
Author(s):  
Misuk Kang ◽  
Ka Young Chung ◽  
Jeffery W. Walker

Catecholamines, endothelin-1 and angiotensin II are among a diverse group of diffusible extracellular signals that regulate pump function of the heart by binding to G-protein coupled receptors (GPCR). When the body demands a temporary boost of power output or if temporary budgeting of resources is required, these signals can adjust heart rate and contractile strength to maintain continuous perfusion of all vascular beds with nutrient- and oxygen-rich blood. Given adequate time in the face of prolonged challenges, activation of GPCRs can also promote “remodeling of the heart” by increasing cell size, organ size, and chamber dimensions, or by varying tissue composition and altering the expression of protein isoforms controlling excitability and contractility. A common feature of heart disease is the state of chronic activation of GPCR signaling systems. Paradoxically, whereas acute activation is beneficial, chronic activation often contributes to further deterioration of cardiac performance. A better understanding of how chronic GPCR activation contributes to the development of heart disease is needed so that it can be translated into better prevention and therapeutic strategies in the clinic.


Sign in / Sign up

Export Citation Format

Share Document