Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurosci ; 32(42): 14709-21, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-23077056

ABSTRACT

The number of functional transient receptor potential vanilloid 1 (TRPV1) channels at the surface, especially at the peripheral terminals of primary sensory neurons, regulates heat sensitivity, and increased surface localization of TRPV1s contributes to heat hyperalgesia. However, the mechanisms for regulating TRPV1 surface localization are essentially unknown. Here, we show that cyclin-dependent kinase 5 (Cdk5), a new player in thermal pain sensation, positively regulates TRPV1 surface localization. Active Cdk5 was found to promote TRPV1 anterograde transport in vivo, suggesting a regulatory role of Cdk5 in TRPV1 membrane trafficking. TRPV1-containing vesicles bind to the forkhead-associated (FHA) domain of the KIF13B (kinesin-3 family member 13B) and are thus delivered to the cell surface. Overexpression of Cdk5 or its activator p35 promoted and inhibition of Cdk5 activity prevented the KIF13B-TRPV1 association, indicating that Cdk5 promotes TRPV1 anterograde transport by mediating the motor-cargo association. Cdk5 phosphorylates KIF13B at Thr-506, a residue located in the FHA domain. T506A mutation reduced the motor-cargo interaction and the cell-permeable TAT-T506 peptide, targeting to the Thr-506, decreased TRPV1 surface localization, demonstrating the essential role of Thr-506 phosphorylation in TRPV1 transport. Moreover, complete Freund's adjuvant (CFA) injection-induced activation of Cdk5 increased the anterograde transport of TRPV1s, contributing to the development and possibly the maintenance of heat hyperalgesia, whereas intrathecal delivery of the TAT-T506 peptide alleviated CFA-induced heat hyperalgesia in rats. Thus, Cdk5 regulation of TRPV1 membrane trafficking is a fundamental mechanism controlling the heat sensitivity of nociceptors, and moderate inhibition of Thr-506 phosphorylation during inflammation might be helpful for the treatment of inflammatory thermal pain.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Hot Temperature/adverse effects , Kinesins/physiology , Membrane Glycoproteins/metabolism , Nociceptors/metabolism , Pain Threshold/physiology , TRPV Cation Channels/metabolism , Animals , Animals, Newborn , CHO Cells , Cell Line, Tumor , Cells, Cultured , Cricetinae , Cricetulus , Male , Phosphorylation , Protein Binding/physiology , Protein Structure, Tertiary , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/antagonists & inhibitors , Threonine/metabolism
2.
J Neurosci ; 29(11): 3551-64, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19295160

ABSTRACT

Morphine is the most commonly used and most effective analgesic in the clinic. However, its use is limited by the tolerance. Evidence indicates that the delta-opioid receptor (DOR) is essential for morphine antinociceptive tolerance; however, their underlying mechanisms are poorly understood. Here, we show that cyclin-dependent kinase 5 (Cdk5), activated in morphine antinociceptive tolerance, directly phosphorylates DOR at Thr-161 in DRG neurons. Cdk5 was found to phosphorylate Thr-161 in the second loop of DOR, but not the corresponding residue in the mu-opioid receptor (MOR). Phosphorylation at Thr-161 is required for normal cell surface expression of DOR, and the formation of DOR-MOR heterodimers. Our studies indicated that inhibition of Cdk5 activity or overexpression of a DOR mutant lacking the Cdk5 phosphorylation site displayed relatively low cell surface expression and relatively low abilities to form heterodimers of DOR and MOR; intrathecal delivery of a construct expressing the T161A mutant of DOR attenuated morphine antinociceptive tolerance in rats, suggesting that Thr-161 phosphorylation of DOR contributed to Cdk5-mediated morphine antinociceptive tolerance. Furthermore, an engineered Tat fusion-interfering peptide corresponding to the second intracellular loop of DOR (Tat-DOR-2L), reduced the cell surface expression of DOR, disrupted the formation of DOR-MOR heterodimers, and significantly attenuated the development of morphine antinociceptive tolerance after intrathecal injection. The present study indicates that Cdk5-mediated phosphorylation of DOR at Thr-161 plays a crucial role in the development of morphine tolerance and suggests the possibility of targeting DOR phosphorylation at Thr-161 to attenuate morphine antinociceptive tolerance during pain management.


Subject(s)
Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/metabolism , Drug Tolerance/physiology , Morphine/pharmacology , Pain Measurement/methods , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, delta/metabolism , Threonine/metabolism , Amino Acid Sequence , Analgesics, Opioid/pharmacology , Animals , Cell Line, Tumor , Cells, Cultured , Cyclin-Dependent Kinase 5/genetics , Humans , Male , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Pain Measurement/drug effects , Phosphorylation/drug effects , Phosphorylation/genetics , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/genetics , Threonine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...