Elsevier

Neuroscience

Volume 154, Issue 3, 26 June 2008, Pages 1067-1076
Neuroscience

Pain mechanism
Contribution of transient receptor potential vanilloid subfamily 1 to endothelin-1-induced thermal hyperalgesia

https://doi.org/10.1016/j.neuroscience.2008.04.010Get rights and content

Abstract

Endothelin-1 (ET-1) plays an important role in peripheral pain processing. However, the mechanisms of the nociceptive action of ET-1 have not been fully elucidated. In this study, we investigated the contribution of transient receptor potential vanilloid subfamily 1 (TRPV1) to ET-1-induced thermal hyperalgesia. Intraplantar ET-1-induced thermal hyperalgesia was examined by assessing the paw withdrawal latency to noxious heat stimuli. In electrophysiological study, whole-cell patch-clamp recordings were performed to investigate the interaction of ET-1 and TRPV1 using human embryonic kidney 293 (HEK293) cells expressing endothelin type A receptor (ETA) and TRPV1. Intraplantar ET-1 (3, 10 and 30 pmol) produced thermal hyperalgesia in a dose-dependent manner. Thermal hyperalgesia was attenuated by the inhibition of ETA and protein kinase C (PKC) but not that of ETB. ET-1-induced thermal hyperalgesia was significantly attenuated in TRPV1-deficient mice compared with that in wild-type mice. In voltage-clamp experiments, 10 nM capsaicin evoked small inward currents in HEK293 cells expressing TRPV1 and ETA. In the presence of ET-1, capsaicin produced much larger current responses (P<0.05). Mutation at PKC-specific TRPV1 phosphorylation sites (S800A/S502A) and PKC inhibitors inhibited the potentiating effect of ET-1. In addition, ET-1 decreased the temperature threshold for TRPV1 activation in a PKC-dependent manner (from 41.0±0.4 °C to 32.6±0.6 °C). In addition, Western blot analysis was also performed to confirm ET-1-induced phosphorylation of TRPV1. Incubation of ET-1 and intraplantar ET-1 evoked phosphorylation of TRPV1 in HEK293 cells expressing TRPV1 and ETA and the skin, respectively. These results suggest that the sensitization of TRPV1 activity through an ETA-PKC pathway contributes to ET-1-induced thermal hyperalgesia.

Section snippets

Animals

Male C57BL/6-strain mice (wild-type mice, WT mice) (weighing 20–25 g, Japan SLC; Hamamatsu, Shizuoka, Japan) and male TRPV1-deficient mice (TRPV1 KO mice) (weighing 20–25 g, Jackson Laboratory, Bar Harbor, MA, USA) were used for behavioral analyses. Each mouse was used in only one experiment. The mice were housed in a temperature-controlled (21±1 °C) room with a 12-h light/dark cycle and given free access to food and water. All procedures were approved by the Sapporo Medical University Animal

Intraplantar ET-1-induced thermal hyperalgesia in WT mice

Intraplantar ET-1 evoked thermal hyperalgesia as previously reported (Menendez et al., 2003). Although the vehicle did not affect the withdrawal latency, ET-1 (3, 10 and 30 pmol) decreased the withdrawal latencies in a dose-dependent manner (P<0.05) (Fig. 1A,Fig. 2B). Peak effects were observed 10–20 min after ET-1 injection, and the decreased withdrawal latencies returned to the basal withdrawal latencies within 60 min after injection. In order to clarify the involvement of receptor subtypes,

Discussion

Our major finding in the behavioral study was that the activation of PKC and TRPV1 as well as that of ETA contributed to ET-1-induced thermal hyperalgesia. In addition, using a specific antibody, we obtained in vivo and in vitro evidence that ET-1 phosphorylated TRPV1. In an electrophysiological study, ET-1 enhanced TRPV1 responsiveness to capsaicin and decreased the temperature threshold of TRPV1 through PKC activation. Although a recent study using HEK293 cells expressing ETA and TRPV1 or

Conclusion

Our results indicate that the sensitization of TRPV1 activity through an ETA–PKC pathway contributes to ET-1-induced thermal hyperalgesia. Therefore, a strategy to target TRPV1 may be more effective to reduce pain associated with ET-1.

Acknowledgments

T.K. and J.Y. contributed equally to this work. Support was provided by a Grant-in-Aid for Scientific Research (19659403 and 19791076) to T. Kawamata and Y. Niiyama, respectively.

References (37)

  • M. Tominaga et al.

    The cloned capsaicin receptor integrates multiple pain-producing stimuli

    Neuron

    (1998)
  • H. Yamamoto et al.

    Endothelin-1 enhances capsaicin-evoked intracellular Ca2+ response via activation of endothelin A receptor in a protein kinase C epsilon-dependent manner in dorsal root ganglion neurons

    Neuroscience

    (2006)
  • J. Yamamoto et al.

    Down-regulation of mu opioid receptor expression within distinct subpopulations of dorsal root ganglion neurons in a murine model of bone cancer pain

    Neuroscience

    (2008)
  • A. Baamonde et al.

    Involvement of endogenous endothelins in thermal and mechanical inflammatory hyperalgesia in mice

    Naunyn Schmiedebergs Arch Pharmacol

    (2004)
  • M.D. Baker

    Protein kinase C mediates up-regulation of tetrodotoxin-resistant, persistent Na+ current in rat and mouse sensory neurones

    J Physiol

    (2005)
  • G. Bhave et al.

    Protein kinase C phosphorylation sensitizes but does not activate the capsaicin receptor transient receptor potential vanilloid 1 (TRPV1)

    Proc Natl Acad Sci U S A

    (2003)
  • M.J. Caterina et al.

    Impaired nociception and pain sensation in mice lacking the capsaicin receptor

    Science

    (2000)
  • B. Dahlof et al.

    Regional haemodynamic effects of endothelin-1 in rat and man: unexpected adverse reaction

    J Hypertens

    (1990)
  • Cited by (54)

    • Mechanisms involved in facial heat hyperalgesia induced by endothelin-1 in female rats

      2017, Archives of Oral Biology
      Citation Excerpt :

      In the dorsal root ganglia (DRG) neurons, it has been reported that ETA receptors are widely expressed in TRPV1-positive small diameter sensory neurons, while expression of ETB receptors is restricted to satellite glial cells (Plant et al., 2007; Plant, Zollner, Mousa, & Oksche, 2006; Pomonis, Rogers, Peters, Ghilardi, & Mantyh, 2001). ET-1 application promotes large increases in capsaicin-evoked currents on mice and rats DRG neurons, an effect that seems to be mediated by ETA receptors and to involve the activation of protein kinase C (PKC) (Kawamata et al., 2008; Plant et al., 2006; Plant et al., 2007). Corroborating these in vitro observations, it was reported that ET-1 induced heat hyperalgesia was attenuated in TRPV1-deficient mice (Ji et al., 2007; Kawamata et al., 2008).

    • A Mechanistic Approach to the Development of Gene Therapy for Chronic Pain

      2016, International Review of Cell and Molecular Biology
    View all citing articles on Scopus
    View full text