Phosphoinositide-3-kinase and mitogen activated protein kinase signaling pathways mediate acute NGF sensitization of TRPV1

https://doi.org/10.1016/j.mcn.2007.01.005Get rights and content

Abstract

Nerve growth factor (NGF) induces an acute sensitization of nociceptive DRG neurons, in part, through sensitization of the capsaicin receptor TRPV1 via the high affinity trkA receptor. The mechanisms linking trkA and TRPV1 remain controversial with several candidate signaling pathways proposed. Utilizing adult rat and mouse DRG neurons and CHO cells co-expressing trkA and TRPV1, we have investigated the signaling events underlying acute TRPV1 sensitization by NGF combining biochemical, electrophysiological, pharmacological, mutational and genetic knockout approaches. Pharmacological interference with p42/p44 mitogen activated protein kinase (MAPK) or phosphoinositide-3-kinase (PI3K), but not PLC abrogated sensitization of capsaicin responses. Co-expression of TRPV1 with wild-type or Y785F (PLC signal deficient) mutant human trkA reconstituted NGF sensitization. In contrast, TRPV1 co-expressed with MAPK signaling deficient Y490A or PI3K signaling deficient Y751F trkA mutants exhibited weaker sensitization. Biochemical analysis of p42/p44 and Akt phosphorylation confirmed the specificity of pharmacological agents and trkA mutants. Finally, NGF sensitization of capsaicin responses was greatly reduced in neurons from p85α (regulatory subunit of PI3K) null mice. These data strongly suggest that PI3K and MAPK pathways, but not the PLC pathway underlie the acute sensitization of TRPV1 by NGF.

Introduction

The transient receptor potential vanilloid 1 (TRPV1) receptor, a member of the transient receptor potential superfamily of cation channels, was expression cloned as the capsaicin receptor (Caterina et al., 1997). TRPV1 is highly expressed in small diameter sensory neurons including those in dorsal root ganglia (DRG) and trigeminal ganglia (TG). Its activation by different noxious stimuli including heat, protons and chemicals such as capsaicin and anandamide, as well as the striking similarities between the behavior of the cloned TRPV1 and the native receptor expressed in DRG or TG neurons (Tominaga et al., 1998), have established it as a molecular marker of nociceptive sensory neurons. Several molecules released during inflammation or injury, including histamine, protons, bradykinin, prostaglandins and nerve growth factor (NGF), can induce hyperalgesia or allodynia, primarily through sensitization of primary nociceptors. Evidence has accumulated that several of these factors including bradykinin and prostaglandins can trigger this sensitizing effect on sensory neurons through protein kinase C (PKC) or cAMP-dependent protein kinase (PKA)-mediated phosphorylation of TRPV1 (Premkumar and Ahern, 2000, Chuang et al., 2001, Sugiuar et al., 2004, Lopshire and Nicol, 1998, Bhave et al., 2002, Bhave et al., 2003, Mohapatra and Nau, 2003). The details of the mechanisms by which other agents including NGF induce sensitization remain unclear and controversial (Zhang and McNaughton, 2006).

It is well documented that NGF plays a critical role in the development and survival of primary nociceptors through transcriptional mechanisms. However, recent studies have shown that NGF is also an important factor in inflammatory or injury-induced hyperalgesia. NGF can promote maintenance of hyperalgesic states through p38 MAP-kinase-mediated non-transcriptional increases in TRPV1 expression in skin (Ji et al., 2002). In addition to such long-term affects, acute sensitization of sensory neurons by NGF has been observed. In cultured rat DRG neurons NGF can increase TTX-resistant sodium currents and reduce voltage-gated potassium currents, effects attributed to NGF signaling through the low affinity p75 receptor (Zhang et al., 2002). Shu and Mendell, 1999a, Shu and Mendell, 1999b first reported an acute sensitization of capsaicin responses by NGF in acutely dissociated rat DRG neurons, a response that can be recapitulated by co-expression of the high affinity trkA receptor and TRPV1 in heterologous systems (Chuang et al., 2001, Zhu and Oxford, 2003, Zhu et al., 2004). Moreover, there is a developmental switch in acute NGF sensitization of TRPV1 in postnatal rat DRG neurons (Zhu et al., 2004), which likely reflects plasticity in the signaling pathway linking the two receptors rather than changes in expression of either receptor. TrkA, as a receptor tyrosine kinase, traditionally signals through activation of one of three major biochemical pathways: phospholipase C (PLCγ), p42/p44 mitogen-activated protein kinase (ERK) or phosphoinositide-3-kinase (PI3K). Although several studies have attempted to elucidate the molecular mechanisms through which NGF sensitizes TRPV1, a consensus has not yet been achieved as published studies have utilized different cell types, different functional endpoints, and have not routinely verified biochemical specificity of signaling interventions.

Using the Xenopus oocyte expression system, the Julius laboratory first presented evidence suggesting that NGF-TrkA activation of PLCγ and subsequent hydrolysis of phosphatidylinositol-4,5-biphosphate (PIP2) relieves a tonic inhibition of TRPV1 by PIP2 (Chuang et al., 2001). A subsequent study identified a C-terminal domain in TRPV1 between amino acids 777 and 820 critical for the NGF sensitization (Prescott and Julius, 2003). PLCγ was also suggested to play a role in the NGF-induced sensitization of heat responses in adult DRG neurons (Galoyan et al., 2003). However, a recent study using calcium imaging of cultured mouse DRG neurons has suggested the sensitizing effect of NGF involves the PI3K pathway, but not the ERK or PLCγ pathways (Bonnington and McNaughton, 2003). An additional study in primary adult rat DRG neurons has suggested that both PI3K and ERK mediate inflammatory heat hyperalgesia through TRPV1 sensitization (Zhuang et al., 2004). Similarly our preliminary investigations in both adult rat DRG neurons and in Chinese Hamster Ovary (CHO) cells co-expressing TRPV1 and TrkA support the involvement of both PI3K and ERK pathways, but not the PLCγ pathway, in acute sensitization of TRPV1 by NGF (Zhu and Oxford, 2003). Furthermore, it has been recently reported that PIP2 resynthesis is necessary for recovery of functional TRPV1 following desensitization (Liu et al., 2005), an observation seemingly opposite to the prediction of an inhibitory role for PIP2. Finally, two recent reports suggest that the critical endpoint of acute trkA signaling in DRG neurons is enhanced trafficking of TRPV1 to the plasma membrane perhaps involving phosphorylation of TRPV1 by src kinase (Zhang et al., 2005, Stein et al., 2006).

To further elucidate the complicated signaling mechanisms between trkA receptors and TRPV1 sensitization, we have combined biochemical, mutagenesis and electrophysiological approaches in both rat and mouse neurons and in mammalian expression systems. Our results indicate that NGF sensitizes TRPV1 independent of extracellular calcium, that this sensitizing effect is mediated by TrkA and requires its downstream PI3K and ERK pathways, possibly in part through activation of ERK by PI3K. While it appears unlikely that PLCγ hydrolysis of PIP2 is directly involved in this phenomenon, a related signaling pathway through protein kinase C (PKC) modulates NGF-induced TRPV1 sensitization. Finally, src kinase appears to be only one of the final effectors involved but cannot account for TRPV1 sensitization in its entirety.

Section snippets

NGF sensitizes capsaicin responses in adult rat DRG neurons through the TrkA receptor

NGF can acutely enhance TRPV1 responses in rat DRG neurons in part by abrogating the normal desensitization that occurs with repeated capsaicin applications (Koplas et al., 1997, Shu and Mendell, 1999a, Shu and Mendell, 1999b, Zhu et al., 2004). To confirm that NGF can additionally sensitize TRPV1, whole cell currents from cultured (24 h with no NGF) adult rat DRG neurons were recorded using standard patch clamp techniques in a bath solution in which no extracellular calcium was added and with

Discussion

TRPV1 is non-specific cation channel initially cloned as the capsaicin receptor and exhibiting activation or modulation by a variety of nociceptive stimuli including heat, protons and chemicals such as capsaicin. It is believed that sensitization and desensitization of TRPV1 are particularly important molecular features correlated with pathologic hyperalgesia and therapeutic hypoalgesia, respectively. Many studies have demonstrated that hyperalgesia can be caused by factors released during

Animals and reagents

Male adult Sprague–Dawley rats (150–180 g) and adult p85α−/− mice were used under Indiana University School of Medicine Laboratory Animal Resource Center Guidelines. The animal room was artificially illuminated from 7:00 A.M. to 7:00 P.M. Capsaicin was purchased from Sigma (St. Louis, MO). NGF was purchased from Alomone Labs (Jerusalem, Israel). The TrkA inhibitor K252a, the MEK (ERK kinase) inhibitors PD98059 and U0126, the PI3K inhibitors LY294002 and wortmannin, the PLC inhibitor U73122 and

Acknowledgments

The authors express their appreciation to Alice Russell for expert assistance in preparation of DRG cultures, to David Julius for the TRPV1 plasmid, to Moses Chao for his gift of human TrkA plasmids, to Reuben Kapur for providing p85α−/− mice, and to Lorne Mendell, Michael Vasko, Grant Nicol and Ted Cummins for helpful discussions during the course of this work. The authors would also like to thank Sharona Gordon and colleagues for sharing their data on PI3K binding to TRPV1 in advance of

References (37)

  • M.J. Caterina et al.

    The capsaicin receptor: a heat-activated ion channel in the pain pathway

    Nature

    (1997)
  • H.H. Chuang et al.

    Bradykinin and nerve growth factor release the capsaicin receptor from PtdIns(4,5)P2-mediated inhibition

    Nature

    (2001)
  • J. Ferreira et al.

    Contribution of vanilloid receptors to the overt nociception induced by B2 kinin receptor activation in mice

    Br. J. Pharmacol.

    (2004)
  • S.M. Galoyan et al.

    Mechanisms of sensitization of the response of single dorsal root ganglion cells from adult rat to noxious heat

    Eur. J. Neurosci.

    (2003)
  • R. Hardie

    Regulation of TRP channels by lipid second messengers

    Annu. Rev. Physiol.

    (2003)
  • H.J. Hu et al.

    Prostaglandin and protein kinase A-dependent modulation of vanilloid receptor function by metabotropic glutamate receptor 5: potential mechanism for thermal hyperalgesia

    J. Neurosci.

    (2002)
  • P.A. Koplas et al.

    The role of calcium in the desensitization of capsaicin responses in rat dorsal root ganglion neurons

    J. Neurosci.

    (1997)
  • B. Liu et al.

    Functional recovery from desensitization of vanilloid receptor TRPV1 requires resynthesis of phosphatidylinositol 4,5-bisphosphate

    J. Neurosci.

    (2005)
  • Cited by (144)

    • 5.10 - Neurotrophins and Pain

      2020, The Senses: A Comprehensive Reference: Volume 1-7, Second Edition
    View all citing articles on Scopus
    View full text