Investigation of TRPV1 loss-of-function phenotypes in transgenic shRNA expressing and knockout mice

https://doi.org/10.1016/j.mcn.2007.12.006Get rights and content

Abstract

The function of the transient receptor potential vanilloid 1 (TRPV1) cation channel was analyzed with RNA interference technologies and compared to TRPV1 knockout mice. Expression of shRNAs targeting TRPV1 in transgenic (tg) mice was proven by RNase protection assays, and TRPV1 downregulation was confirmed by reduced expression of TRPV1 mRNA and lack of receptor agonist binding in spinal cord membranes. Unexpectedly, TRPV3 mRNA expression was upregulated in shRNAtg but downregulated in knockout mice. Capsaicin-induced [Ca2+]i changes in small diameter DRG neurons were significantly diminished in TRPV1 shRNAtg mice, and administration of capsaicin hardly induced hypothermia or nocifensive behaviour in vivo. Likewise, sensitivity towards noxious heat was reduced. Interestingly, spinal nerve injured TRPV1 knockout but not shRNAtg animals developed mechanical allodynia and hypersensitivity. The present study provides further evidence for the relevance of TRPV1 in neuropathic pain and characterizes RNA interference as valuable technique for drug target validation in pain research.

Introduction

TRPV1 is a cation channel that is predominantly expressed by nociceptive sensory neurons. TRPV1 is activated by capsaicin, noxious heat and protons (Caterina et al., 1997, Tominaga et al., 1998). A variety of intracellular pain-related pathways and molecules regulate nociception and signal transmission by TRPV1 (Szallasi et al., 2007, Tominaga, 2007). As TRPV1 appears to be a central molecular integrator of noxious stimuli, it is considered to be an attractive target for new analgesic drugs.

Knockout mice lacking TRPV1 exhibit diminished sensitivity to heat and perturbed micturition (Birder et al., 2002, Caterina et al., 2000, Davis et al., 2000). Interestingly, TRPV1 knockout mice do not reveal a significantly altered phenotype in the partial sciatic nerve ligation model of mononeuropathic pain as compared to wild-type animals (Caterina et al., 2000) and even show increased mechanical hyperalgesia in polyneuropathic pain models (Bolcskei et al., 2005). In contrast, intrathecal administration of the TRPV1 antagonist capsazepine was reported to block A-fiber-evoked responses in dorsal horn neurons of rats after spinal nerve ligation (Kelly and Chapman, 2002), and more selective TRPV1 antagonists attenuated mechanical allodynia and hyperalgesia in rat models of mononeuropathic pain (Christoph et al., 2006, Honore et al., 2005, Kanai et al., 2005, Pomonis et al., 2003). The precise role of TRPV1 in neuropathic pain is therefore still controversial.

We decided to use an RNA interference (RNAi) approach to investigate further the role of TRPV1 in an animal model of neuropathic pain. RNAi is an evolutionary conserved mechanism of posttranscriptional gene silencing mediated by double stranded RNA molecules (Grünweller and Hartmann, 2005, Kim and Rossi, 2007). For applications in mammalian cells, 21 nucleotides long small interfering RNAs (siRNAs) can be employed to specifically silence a particular gene. To achieve long-term inhibition of a target gene, self-complementary short hairpin (shRNAs) can be expressed intracellularly (Shi, 2003). The shRNAs are processed by the RNase Dicer to give siRNA-type molecules. For in vivo experiments, transgenic animals have been generated which continuously express the shRNA, thereby permanently knocking down the targeted gene (Seibler et al., 2005).

Several studies have already demonstrated the potential of RNAi to validate new targets in pain research (for reviews, see Ganju and Hall, 2004, Röhl and Kurreck, 2006). The pain-related RNAi approaches published to date made use of repeated injections or continuous infusion of siRNA molecules. With this experimental design, however, the knockdown was intended to last only for a limited time of several days. We have also performed some initial functional investigations by transient inhibition of TRPV1 expression by intrathecal injection of antisense oligonucleotides (Christoph et al., 2007) or siRNA molecules (Christoph et al., 2006).

In order to perform a detailed comparison of different gene silencing strategies, we generated transgenic animals that express a TRPV1-directed shRNA and analyzed expression of the transgene, knockdown of TRPV1 and pain-related behaviour. Our study demonstrates that transgenic animals expressing shRNAs can be used for functional investigations in pain research and suggests a role of TRPV1 in neuropathic pain.

Section snippets

Generation of shRNA transgenic mice

In a previous study, we screened for efficient siRNAs targeting the mRNA of TRPV1. One siRNA, named VsiR1, was found to inhibit expression of a TRPV1–GFP fusion construct in the subnanomolar range in co-transfection experiments (Grünweller et al., 2003) and was efficacious upon intrathecal administration in rodent models of mononeuropathic and visceral pain (Christoph et al., 2006). In vitro and in vivo activities of this particular siRNA have recently been confirmed by an independent group (

Discussion

The present study successfully demonstrates that RNAi is a useful approach to validate potential novel targets for pain therapy. Previous studies have shown that the knockdown of the expression of several pain-related genes, such as the delta opioid receptor (Luo et al., 2005), the ionotropic nucleotide receptor P2X3 (Dorn et al., 2004), the NMDA receptor NR2B subunit (Tan et al., 2005) or TRPV1 (Christoph et al., 2006), by acute or sub-acute local administration of siRNAs resulted in analgesic

Construction and testing of the shRNA expression vector

For the generation of transgenic animals, an exchange vector, named pRMCE-U6-shTrpv1, was constructed that encodes an shRNA under control of the U6 promotor: The vector contains the F3 site and the FRT site in the same configuration as in the rosa26 targeting vector described by Seibler et al. (2005). The vector was generated using standard cloning procedures and has the following order in 5′ to 3′ direction: synthetic polyA signal, F3 site and a neomycin resistance gene lacking the start ATG.

Acknowledgments

The technical assistance of Birgit Bieber, Franz-Josef Butz, Elisabeth Haase, Elisabeth Krings, Thomas Krüger, Beatrice Petter, Simone Pfennings, Silke Rosenow, Stefanie Voß and the secretarial assistance of Ingrid Loeser is gratefully acknowledged. Furthermore, we want to thank Clemens Gillen, Kurexi Yunusi and Arnold Grünweller for initial contributions to the project and Oliver Bogen and Frank Nicolai Single for helpful discussion. The work was sponsored by Grünenthal GmbH, RiNA network for

References (58)

  • E.N. Olson et al.

    Know your neighbors: three phenotypes in null mutants of the myogenic bHLH gene MRF4

    Cell

    (1996)
  • M.H. Ossipov et al.

    Lack of involvement of capsaicin-sensitive primary afferents in nerve-ligation injury induced tactile allodynia in rats

    Pain

    (1999)
  • M. Petersen et al.

    Relationships between capsaicin sensitivity of mammalian sensory neurons, cell size and type of voltage gated Ca-currents

    Brain Res.

    (1991)
  • T. Sakurada et al.

    The capsaicin test in mice for evaluating tachykinin antagonists in the spinal cord

    Neuropharmacology

    (1992)
  • Y. Shi

    Mammalian RNAi for the masses

    Trends Genet.

    (2003)
  • M. Tominaga et al.

    The cloned capsaicin receptor integrates multiple pain-producing stimuli

    Neuron

    (1998)
  • M. Zimmermann

    Ethical guidelines for investigations of experimental pain in conscious animals

    Pain

    (1983)
  • H.H. Arnold et al.

    Targeted inactivation of myogenic factor genes reveals their role during mouse myogenesis: a review

    Int. J. Dev. Biol.

    (1996)
  • L.A. Birder et al.

    Altered urinary bladder function in mice lacking the vanilloid receptor TRPV1

    Nat. Neurosci.

    (2002)
  • A. Birmingham et al.

    3′ UTR seed matches, but not overall identity, are associated with RNAi off-targets

    Nat. Methods

    (2006)
  • M.J. Caterina et al.

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

    Nature

    (1997)
  • M.J. Caterina et al.

    Impaired nociception and pain sensation in mice lacking the capsaicin receptor

    Science

    (2000)
  • J.B. Davis et al.

    Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia

    Nature

    (2000)
  • A.T. De Souza et al.

    Transcriptional and phenotypic comparisons of Ppara knockout and siRNA knockdown mice

    Nucleic Acids Res.

    (2006)
  • G. Dorn et al.

    siRNA relieves chronic neuropathic pain

    Nucleic Acids Res.

    (2004)
  • N.B. Eddy et al.

    Synthetic analgesics. II. Dithienylbutenyl- and dithienylbutylamines

    J. Pharmacol. Exp. Ther.

    (1953)
  • K. Eggan et al.

    Hybrid vigor, fetal overgrowth, and viability of mice derived by nuclear cloning and tetraploid embryo complementation

    Proc. Natl. Acad. Sci. U. S. A.

    (2001)
  • P. Facer et al.

    Differential expression of the capsaicin receptor TRPV1 and related novel receptors TRPV3, TRPV4 and TRPM8 in normal human tissues and changes in traumatic and diabetic neuropathy

    BMC Neurol.

    (2007)
  • P. Ganju et al.

    Potential applications of siRNA for pain therapy

    Expert Opin. Biol. Ther.

    (2004)
  • Cited by (0)

    View full text