Endothelin potentiates TRPV1 via ETA receptor-mediated activation of protein kinase C.

Standard

Endothelin potentiates TRPV1 via ETA receptor-mediated activation of protein kinase C. / Plant, Tim D; Zöllner, Christian; Kepura, Frauke; Mousa, Shaaban S; Eichhorst, Jenny; Schaefer, Michael; Furkert, Jens; Stein, Christoph; Oksche, Alexander.

In: MOL PAIN, Vol. 3, 2007, p. 35.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Plant, TD, Zöllner, C, Kepura, F, Mousa, SS, Eichhorst, J, Schaefer, M, Furkert, J, Stein, C & Oksche, A 2007, 'Endothelin potentiates TRPV1 via ETA receptor-mediated activation of protein kinase C.', MOL PAIN, vol. 3, pp. 35. <http://www.ncbi.nlm.nih.gov/pubmed/18001466?dopt=Citation>

APA

Plant, T. D., Zöllner, C., Kepura, F., Mousa, S. S., Eichhorst, J., Schaefer, M., Furkert, J., Stein, C., & Oksche, A. (2007). Endothelin potentiates TRPV1 via ETA receptor-mediated activation of protein kinase C. MOL PAIN, 3, 35. http://www.ncbi.nlm.nih.gov/pubmed/18001466?dopt=Citation

Vancouver

Plant TD, Zöllner C, Kepura F, Mousa SS, Eichhorst J, Schaefer M et al. Endothelin potentiates TRPV1 via ETA receptor-mediated activation of protein kinase C. MOL PAIN. 2007;3:35.

Bibtex

@article{09e77e76cb234355ad9a7d5721caa030,
title = "Endothelin potentiates TRPV1 via ETA receptor-mediated activation of protein kinase C.",
abstract = "BACKGROUND: Endothelin-1 (ET-1) both stimulates nociceptors and sensitizes them to noxious stimuli, an effect probably mediated by the ETA receptor (ETAR) expressed in sensory neurons. The cellular mechanisms of this ET-1-mediated effect are only poorly understood. TRPV1, the heat-, pH- and capsaicin-sensitive cation channel already known to be modulated by a number of cellular mediators released in response to noxious stimuli and during inflammation, is a potential target for the action of ET-1. RESULTS: We studied the effects of ET-1 on TRPV1 in sensory neurons from the dorsal root ganglion (DRG) and in HEK293 cells coexpressing TRPV1 and the ETAR. Specific 125I-ET-1 binding sites (817 +/- 92 fmol/mg) were detected in membrane preparations of DRG with an ETAR/ETBR ratio of 60:40. In an immunofluorescence analysis, coexpression of TRPV1 and the ETAR was found in a subpopulation of primary sensory neurons. ET-1 strongly potentiated capsaicin-induced TRPV1 currents in some neurons, and in HEK293 cells co-expressing TRPV1 and the ETAR. Weaker potentiation was observed in HEK293 cells coexpressing TRPV1 and the ETBR. ETAR activation also increased responses to low pH and heat. In HEK293 cells, strong potentiation of TRPV1 like that induced by ET-1 via the ETAR could be induced by PKC activation, but not with activators of the adenylyl cyclase or the PKA pathway. Furthermore, inhibition of PKC with bisindolylmaleimide X (BIM X) or mutation of the PKC phosphorylation site S800 completely prevented ETAR-mediated potentiation. CONCLUSION: We conclude that ET-1 potentiates TRPV1 by a PKC-dependent mechanism and that this could play a major role in the algogenic and hyperalgesic effects of ET-1 described in previous studies.",
author = "Plant, {Tim D} and Christian Z{\"o}llner and Frauke Kepura and Mousa, {Shaaban S} and Jenny Eichhorst and Michael Schaefer and Jens Furkert and Christoph Stein and Alexander Oksche",
year = "2007",
language = "Deutsch",
volume = "3",
pages = "35",

}

RIS

TY - JOUR

T1 - Endothelin potentiates TRPV1 via ETA receptor-mediated activation of protein kinase C.

AU - Plant, Tim D

AU - Zöllner, Christian

AU - Kepura, Frauke

AU - Mousa, Shaaban S

AU - Eichhorst, Jenny

AU - Schaefer, Michael

AU - Furkert, Jens

AU - Stein, Christoph

AU - Oksche, Alexander

PY - 2007

Y1 - 2007

N2 - BACKGROUND: Endothelin-1 (ET-1) both stimulates nociceptors and sensitizes them to noxious stimuli, an effect probably mediated by the ETA receptor (ETAR) expressed in sensory neurons. The cellular mechanisms of this ET-1-mediated effect are only poorly understood. TRPV1, the heat-, pH- and capsaicin-sensitive cation channel already known to be modulated by a number of cellular mediators released in response to noxious stimuli and during inflammation, is a potential target for the action of ET-1. RESULTS: We studied the effects of ET-1 on TRPV1 in sensory neurons from the dorsal root ganglion (DRG) and in HEK293 cells coexpressing TRPV1 and the ETAR. Specific 125I-ET-1 binding sites (817 +/- 92 fmol/mg) were detected in membrane preparations of DRG with an ETAR/ETBR ratio of 60:40. In an immunofluorescence analysis, coexpression of TRPV1 and the ETAR was found in a subpopulation of primary sensory neurons. ET-1 strongly potentiated capsaicin-induced TRPV1 currents in some neurons, and in HEK293 cells co-expressing TRPV1 and the ETAR. Weaker potentiation was observed in HEK293 cells coexpressing TRPV1 and the ETBR. ETAR activation also increased responses to low pH and heat. In HEK293 cells, strong potentiation of TRPV1 like that induced by ET-1 via the ETAR could be induced by PKC activation, but not with activators of the adenylyl cyclase or the PKA pathway. Furthermore, inhibition of PKC with bisindolylmaleimide X (BIM X) or mutation of the PKC phosphorylation site S800 completely prevented ETAR-mediated potentiation. CONCLUSION: We conclude that ET-1 potentiates TRPV1 by a PKC-dependent mechanism and that this could play a major role in the algogenic and hyperalgesic effects of ET-1 described in previous studies.

AB - BACKGROUND: Endothelin-1 (ET-1) both stimulates nociceptors and sensitizes them to noxious stimuli, an effect probably mediated by the ETA receptor (ETAR) expressed in sensory neurons. The cellular mechanisms of this ET-1-mediated effect are only poorly understood. TRPV1, the heat-, pH- and capsaicin-sensitive cation channel already known to be modulated by a number of cellular mediators released in response to noxious stimuli and during inflammation, is a potential target for the action of ET-1. RESULTS: We studied the effects of ET-1 on TRPV1 in sensory neurons from the dorsal root ganglion (DRG) and in HEK293 cells coexpressing TRPV1 and the ETAR. Specific 125I-ET-1 binding sites (817 +/- 92 fmol/mg) were detected in membrane preparations of DRG with an ETAR/ETBR ratio of 60:40. In an immunofluorescence analysis, coexpression of TRPV1 and the ETAR was found in a subpopulation of primary sensory neurons. ET-1 strongly potentiated capsaicin-induced TRPV1 currents in some neurons, and in HEK293 cells co-expressing TRPV1 and the ETAR. Weaker potentiation was observed in HEK293 cells coexpressing TRPV1 and the ETBR. ETAR activation also increased responses to low pH and heat. In HEK293 cells, strong potentiation of TRPV1 like that induced by ET-1 via the ETAR could be induced by PKC activation, but not with activators of the adenylyl cyclase or the PKA pathway. Furthermore, inhibition of PKC with bisindolylmaleimide X (BIM X) or mutation of the PKC phosphorylation site S800 completely prevented ETAR-mediated potentiation. CONCLUSION: We conclude that ET-1 potentiates TRPV1 by a PKC-dependent mechanism and that this could play a major role in the algogenic and hyperalgesic effects of ET-1 described in previous studies.

M3 - SCORING: Zeitschriftenaufsatz

VL - 3

SP - 35

ER -