scholarly journals Sphingosine 1-Phosphate to p38 Signaling via S1P1 Receptor and Gαi/o Evokes Augmentation of Capsaicin-Induced Ionic Currents in Mouse Sensory Neurons

2014 ◽  
Vol 10 ◽  
pp. 1744-8069-10-74 ◽  
Author(s):  
Michiel Langeslag ◽  
Serena Quarta ◽  
Michael G Leitner ◽  
Michaela Kress ◽  
Norbert Mair
1994 ◽  
Vol 1 (4) ◽  
pp. 243-257
Author(s):  
G A Clark ◽  
R D Hawkins ◽  
E R Kandel

A hallmark of many forms of classical conditioning is a precise temporal specificity: Learning is optimal when the conditioned stimulus (CS) slightly precedes the unconditioned stimulus (US), but the learning is degraded at longer or backward intervals, consistent with the notion that conditioning involves learning about predictive relationships in the environment. To further examine the cellular mechanisms contributing to the temporal specificity of classical conditioning of the siphon-withdrawal response in Aplysia, we paired action potential activity in siphon sensory neurons (the neural CS) with tail nerve shock (the US) at three critical time points. We found that CS-US pairings at short (0.5 sec) forward intervals produced greater synaptic facilitation at sensorimotor connections than did either 0.5-sec backward pairings or longer (5 sec) forward pairings, as reflected in a differential increase in both the amplitude and rate of rise of the synaptic potential. In the same preparations, forward pairings also differentially reduced the sensory neuron afterhyperpolarization relative to backward pairings, suggesting that changes in synaptic efficacy were accompanied by temporally specific changes in ionic currents in the sensory neurons. Additional experiments demonstrated that short forward pairings of sensory cell activity and restricted applications of the neuromodulatory transmitter serotonin (normally released by the US) differentially enhanced action potential broadening in siphon sensory neurons, relative to backward pairings. Taken together, these results suggest that temporally specific synaptic enhancement engages both spike-width-dependent and spike-width-independent facilitatory processes and that activity-dependent enhancement of presynaptic facilitation may contribute to both the CS-US sequence and proximity requirements of conditioning.


2012 ◽  
Vol 22 (8) ◽  
pp. 2794-2797 ◽  
Author(s):  
Qinghua Meng ◽  
Baowei Zhao ◽  
Qiongfeng Xu ◽  
Xuesong Xu ◽  
Guanghui Deng ◽  
...  

Author(s):  
Yanmei Qi ◽  
Norbert Mair ◽  
Kai K. Kummer ◽  
Michael G. Leitner ◽  
María Camprubí-Robles ◽  
...  

2006 ◽  
Vol 96 (3) ◽  
pp. 1042-1052 ◽  
Author(s):  
Y. H. Zhang ◽  
J. C. Fehrenbacher ◽  
M. R. Vasko ◽  
G. D. Nicol

Sphingosine-1-phosphate (S1P) is released by immune cells and is thought to play a key role in chemotaxis and the onset of the inflammatory response. The question remains whether this lipid mediator also contributes to the enhanced sensitivity of nociceptive neurons that is associated with inflammation. Therefore we examined whether S1P alters the excitability of small diameter, capsaicin-sensitive sensory neurons by measuring action potential (AP) firing and two of the membrane currents critical in regulating the properties of the AP. External application of S1P augments the number of APs evoked by a depolarizing current ramp. The enhanced firing is associated with a decrease in the rheobase and an increase in the resistance at firing threshold although neither the firing threshold nor the resting membrane potential are changed. Treatment with S1P enhanced the tetrodotoxin-resistant sodium current and decreased the total outward potassium current ( IK). When sensory neurons were internally perfused with GDP-β-S, a blocker of G protein activation, the S1P-induced increase in APs was completely blocked and suggests the excitatory actions of S1P are mediated through G-protein-coupled receptors called endothelial differentiation gene or S1PR. In contrast, internal perfusion with GDP-β-S and S1P increased the number of APs evoked by the current ramp. These results and our finding that the mRNAs for S1PRs are expressed in both the intact dorsal root ganglion and cultures of adult sensory neurons supports the notion that S1P acts on S1PRs linked to G proteins. Together these findings demonstrate that S1P can regulate the excitability of small diameter sensory neurons by acting as an external paracrine-type ligand through activation of G-protein-coupled receptors and thus may contribute to the hypersensitivity during inflammation.


2006 ◽  
Vol 16 (14) ◽  
pp. 3684-3687 ◽  
Author(s):  
Petr Vachal ◽  
Leslie M. Toth ◽  
Jeffrey J. Hale ◽  
Lin Yan ◽  
Sander G. Mills ◽  
...  

2012 ◽  
Vol 108 (5) ◽  
pp. 1473-1483 ◽  
Author(s):  
Chao Li ◽  
Xian Xuan Chi ◽  
Wenrui Xie ◽  
J. A. Strong ◽  
J.-M. Zhang ◽  
...  

Previously we demonstrated that sphingosine 1-phosphate receptor 1 (S1PR1) played a prominent, but not exclusive, role in enhancing the excitability of small-diameter sensory neurons, suggesting that other S1PRs can modulate neuronal excitability. To examine the potential role of S1PR2 in regulating neuronal excitability we used the established selective antagonist of S1PR2, JTE-013. Here we report that exposure to JTE-013 alone produced a significant increase in excitability in a time- and concentration-dependent manner in 70–80% of recorded neurons. Internal perfusion of sensory neurons with guanosine 5′- O-(2-thiodiphosphate) (GDP-β-S) via the recording pipette inhibited the sensitization produced by JTE-013 as well as prostaglandin E2. Pretreatment with pertussis toxin or the selective S1PR1 antagonist W146 blocked the sensitization produced by JTE-013. These results indicate that JTE-013 might act as an agonist at other G protein-coupled receptors. In neurons that were sensitized by JTE-013, single-cell RT-PCR studies demonstrated that these neurons did not express the mRNA for S1PR2. In behavioral studies, injection of JTE-013 into the rat's hindpaw produced a significant increase in the mechanical sensitivity in the ipsilateral, but not contralateral, paw. Injection of JTE-013 did not affect the withdrawal latency to thermal stimulation. Thus JTE-013 augments neuronal excitability independently of S1PR2 by unknown mechanisms that may involve activation of other G protein-coupled receptors such as S1PR1. Clearly, further studies are warranted to establish the causal nature of this increased sensitivity, and future studies of neuronal function using JTE-013 should be interpreted with caution.


Sign in / Sign up

Export Citation Format

Share Document