Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Piezo2 is the major transducer of mechanical forces for touch sensation in mice

Abstract

The sense of touch provides critical information about our physical environment by transforming mechanical energy into electrical signals1. It is postulated that mechanically activated cation channels initiate touch sensation, but the identity of these molecules in mammals has been elusive2. Piezo2 is a rapidly adapting, mechanically activated ion channel expressed in a subset of sensory neurons of the dorsal root ganglion and in cutaneous mechanoreceptors known as Merkel-cell–neurite complexes3,4. It has been demonstrated that Merkel cells have a role in vertebrate mechanosensation using Piezo2, particularly in shaping the type of current sent by the innervating sensory neuron4,5,6; however, major aspects of touch sensation remain intact without Merkel cell activity4,7. Here we show that mice lacking Piezo2 in both adult sensory neurons and Merkel cells exhibit a profound loss of touch sensation. We precisely localize Piezo2 to the peripheral endings of a broad range of low-threshold mechanoreceptors that innervate both hairy and glabrous skin. Most rapidly adapting, mechanically activated currents in dorsal root ganglion neuronal cultures are absent in Piezo2 conditional knockout mice, and ex vivo skin nerve preparation studies show that the mechanosensitivity of low-threshold mechanoreceptors strongly depends on Piezo2. This cellular phenotype correlates with an unprecedented behavioural phenotype: an almost complete deficit in light-touch sensation in multiple behavioural assays, without affecting other somatosensory functions. Our results highlight that a single ion channel that displays rapidly adapting, mechanically activated currents in vitro is responsible for the mechanosensitivity of most low-threshold mechanoreceptor subtypes involved in innocuous touch sensation. Notably, we find that touch and pain sensation are separable, suggesting that as-yet-unknown mechanically activated ion channel(s) must account for noxious (painful) mechanosensation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Piezo2 is localized at the nerve terminals of sensory neurons that innervate the skin.
Figure 2: Advil-creERT2 mediates efficient deletion of Piezo2 and leads to specific loss of rapidly adapting, mechanically activated currents in cultured DRG neurons.
Figure 3: Piezo2 is required for mechanoreceptor function in ex vivo skin nerve preparation.
Figure 4: Piezo2CKO mice show profound and specific defects in innocuous touch sensation in multiple behavioural assays.

Similar content being viewed by others

References

  1. Abraira, V. E. & Ginty, D. D. The sensory neurons of touch. Neuron 79, 618–639 (2013)

    Article  CAS  PubMed  Google Scholar 

  2. Árnadóttir, J. & Chalfie, M. Eukaryotic mechanosensitive channels. Annu. Rev. Biophys. 39, 111–137 (2010)

    Article  PubMed  Google Scholar 

  3. Coste, B. et al. Piezo1 and Piezo2 are essential components of distinct mechanically activated cation channels. Science 330, 55–60 (2010)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Woo, S.-H. et al. Piezo2 is required for Merkel-cell mechanotransduction. Nature 509, 622–626 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Maksimovic, S. et al. Epidermal Merkel cells are mechanosensory cells that tune mammalian touch receptors. Nature 509, 617–621 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ikeda, R. et al. Merkel cells transduce and encode tactile stimuli to drive Aβ-afferent impulses. Cell 157, 664–675 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Maricich, S. M., Morrison, K. M., Mathes, E. L. & Brewer, B. M. Rodents rely on Merkel cells for texture discrimination tasks. J. Neurosci. 32, 3296–3300 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lechner, S. G. & Lewin, G. R. Hairy sensation. Physiology (Bethesda) 28, 142–150 (2013)

    CAS  Google Scholar 

  9. Maksimovic, S., Baba, Y. & Lumpkin, E. A. Neurotransmitters and synaptic components in the Merkel cell-neurite complex, a gentle-touch receptor. Ann. NY Acad. Sci. 1279, 13–21 (2013)

    Article  ADS  CAS  PubMed  Google Scholar 

  10. Dubin, A. E. et al. Inflammatory signals enhance piezo2-mediated mechanosensitive currents. Cell Rep. 2, 511–517 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Haeberle, H. et al. Molecular profiling reveals synaptic release machinery in Merkel cells. Proc. Natl Acad. Sci. USA 101, 14503–14508 (2004)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lou, S., Duan, B., Vong, L., Lowell, B. B. & Ma, Q. Runx1 controls terminal morphology and mechanosensitivity of VGLUT3-expressing C-mechanoreceptors. J. Neurosci. 33, 870–882 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. McCarter, G. C., Reichling, D. B. & Levine, J. D. Mechanical transduction by rat dorsal root ganglion neurons in vitro. Neurosci. Lett. 273, 179–182 (1999)

    Article  CAS  PubMed  Google Scholar 

  14. Hu, J. & Lewin, G. R. Mechanosensitive currents in the neurites of cultured mouse sensory neurones. J. Physiol. 577, 815–828 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Wetzel, C. et al. A stomatin-domain protein essential for touch sensation in the mouse. Nature 445, 206–209 (2007)

    Article  ADS  CAS  PubMed  Google Scholar 

  16. Moshourab, R. A., Wetzel, C., Martinez-Salgado, C. & Lewin, G. R. Stomatin-domain protein interactions with acid-sensing ion channels modulate nociceptor mechanosensitivity. J. Physiol. 591, 5555–5574 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Maricich, S. M. et al. Merkel cells are essential for light-touch responses. Science 324, 1580–1582 (2009)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wellnitz, S. A., Lesniak, D. R., Gerling, G. J. & Lumpkin, E. A. The regularity of sustained firing reveals two populations of slowly adapting touch receptors in mouse hairy skin. J. Neurophysiol. 103, 3378–3388 (2010)

    Article  PubMed  PubMed Central  Google Scholar 

  19. Rugiero, F., Drew, L. J. & Wood, J. N. Kinetic properties of mechanically activated currents in spinal sensory neurons. J. Physiol. (Lond.) 588, 301–314 (2010)

    Article  CAS  Google Scholar 

  20. Poole, K., Herget, R., Lapatsina, L., Ngo, H. D. & Lewin, G. R. Tuning Piezo ion channels to detect molecular-scale movements relevant for fine touch. Nature Commun. 5, 3520 (2014)

    Article  ADS  Google Scholar 

  21. Petrus, M. et al. A role of TRPA1 in mechanical hyperalgesia is revealed by pharmacological inhibition. Mol. Pain 3, 40 (2007)

    Article  PubMed  PubMed Central  Google Scholar 

  22. Garrison, S. R., Dietrich, A. & Stucky, C. L. TRPC1 contributes to light-touch sensation and mechanical responses in low-threshold cutaneous sensory neurons. J. Neurophysiol. 107, 913–922 (2012)

    Article  CAS  PubMed  Google Scholar 

  23. Dhaka, A. et al. TRPM8 is required for cold sensation in mice. Neuron 54, 371–378 (2007)

    Article  CAS  PubMed  Google Scholar 

  24. Bradbury, E. J. et al. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature 416, 636–640 (2002)

    Article  ADS  CAS  PubMed  Google Scholar 

  25. Eijkelkamp, N. et al. A role for Piezo2 in EPAC1-dependent mechanical allodynia. Nature Commun. 4, 1682 (2013)

    Article  ADS  CAS  Google Scholar 

  26. Ranade, S. S. et al. Piezo1, a mechanically activated ion channel, is required for vascular development in mice. Proc. Natl Acad. Sci. USA 111, 10347–10352 (2014)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Voss, F. K. et al. Identification of LRRC8 heteromers as an essential component of the volume-regulated anion channel VRAC. Science 344, 634–638 (2014)

    Article  ADS  CAS  PubMed  Google Scholar 

  28. Qiu, Z. et al. SWELL1, a plasma membrane protein, is an essential component of volume-regulated anion channel. Cell 157, 447–458 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kim, S. E., Coste, B., Chadha, A., Cook, B. & Patapoutian, A. The role of Drosophila Piezo in mechanical nociception. Nature 483, 209–212 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  30. Faucherre, A., Nargeot, J., Mangoni, M. E. & Jopling, C. piezo2b regulates vertebrate light touch response. J. Neurosci. 33, 17089–17094 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nature Neurosci. 13, 133–140 (2010)

    Article  CAS  PubMed  Google Scholar 

  32. Lau, J. et al. Temporal control of gene deletion in sensory ganglia using a tamoxifen-inducible Advillin-Cre-ERT2 recombinase mouse. Mol. Pain 7, 100 (2011)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods 25, 402–408 (2001)

    CAS  PubMed  Google Scholar 

  34. Kwan, K. Y. et al. TRPA1 contributes to cold, mechanical, and chemical nociception but is not essential for hair-cell transduction. Neuron 50, 277–289 (2006)

    Article  CAS  PubMed  Google Scholar 

  35. Lariviere, W. R. et al. Heritability of nociception. III. Genetic relationships among commonly used assays of nociception and hypersensitivity. Pain 97, 75–86 (2002)

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge T. Goode for help in behavioural analysis. We also thank R. Moran and T. Johnson for assistance with histology, K. Spencer for imaging analysis and M. Braunschweig for technical assistance. S.S.R. was funded by a pre-doctoral fellowship from the California Institute of Regenerative Medicine. R.A.M. was supported by Clinical Research Fellowship from the Max Delbrück Center (MDC). G.R.L.’s laboratory was supported by senior European Research Council grant (project 294678) and a grant from the Deutsche Forschungsgemeinshaft (SFB958 project A9). A.P. is a Howard Hughes Medical Institute Investigator. This study was partly funded by NIH grant R01 DE022358 to A.P.

Author information

Authors and Affiliations

Authors

Contributions

S.S.R. and A.P. designed experiments and wrote the manuscript along with G.R.L. S.S.R., J.N.W. and Z.Q. generated transgenic lines used in this study. S.-H.W. performed all immunostaining experiments. A.E.D. conducted electrophysiology on cultured DRG neurons and J.M. isolated and cultured cells. R.A.M., C.W., V.B. and G.R.L. performed skin nerve electrophysiology and analysed the data. S.S.R., M.P., A.G.F. and K.R. performed behavioural assays on mice. B.C., J.M. and A.J.W. developed the novel two-choice mechanosensory instrument.

Corresponding author

Correspondence to Ardem Patapoutian.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Mechanically activated currents elicited in cultured DRG neurons from Piezo2WT and Piezo2CKO mice by poking with a blunt probe.

a, Representative traces of rapidly adapting (RA) currents in Piezo2WT (top) and Piezo2CKO (bottom). Piezo2WT DRG neurons show characteristic rapidly adapting currents; a subpopulation can be active with apparent low thresholds (right). Piezo2CKO mice contained a few rapidly adapting type cells but none appeared to be low-threshold mechanoreceptors. b, c, Representative traces of intermediately adapting (IA) and slowly adapting (SA) currents, respectively, with no observable differences between the two genotypes. All data were low-pass filtered off line at 4 kHz. Action potentials were elicited by current injection in all neurons. Piezo2WT: RA, left: 20 μm diameter, 5 μm apparent threshold; RA, right: 28 μm diameter, 1 μm apparent threshold; IA: 23 μm diameter, 6 μm apparent threshold; SA: 20 μm diameter; 2 μm apparent threshold. Piezo2CKO: RA, left: 23 μm diameter, 8 μm apparent threshold; RA, right: none found; IA: 20 μm diameter, 5.5 μm apparent threshold; SA: 30 μm diameter, 8 μm apparent threshold. Lower right shows an example of the probe displacement protocol (stimulus). Results from n = 3 independent experiments.

Extended Data Figure 2 Apparent threshold analysis of Piezo2WT and Piezo2CKO DRG neurons.

The smallest soma indentation eliciting a detectable mechanically activated response (apparent threshold) depends, in part, on the incremental distance applied (0.5 μm) and the proportional displacement in relation to the soma diameter. The apparent thresholds of all rapidly adapting responses normalized to soma diameter reveal a wide range of sensitivities of Piezo2WT DRG neurons (black) and the high apparent threshold responses of the remaining rapidly adapting neurons in Piezo2CKO DRG neurons (red). The lowest apparent thresholds are observed only in Piezo2WT. Results from n = 3 independent experiments.

Extended Data Figure 3 Expression of various markers of subpopulations of DRG neurons are similar in Piezo2WT and Piezo2CKO mice.

a, b, Representative images from immunofluorescence of Nefh in DRGs from Piezo2WT (a) or Piezo2CKO (b) mice. c, d, Representative image from immunofluorescence of thymidine hydroxylase (TH) in DRGs from Piezo2WT (c) or Piezo2CKO (d) mice. e, f, Representative image from immunofluorescence of CGRP in DRGs from Piezo2WT (e) or Piezo2CKO (f) mice. All markers stained in green. Scale bars, 100 μm.

Extended Data Figure 4 DRG innervation of skin is unaffected in Piezo2CKO mice.

a, d, Representative image of immunostaining of Krt8 (green) and Nefh (red) in Merkel-cell–neurite complexes in Piezo2WT (a) and Piezo2CKO (d) glabrous skin. b, e, f, Representative image of immunostaining of S100 (green) and Nefh (red) in circumferential fibres (arrowheads) and lanceolate endings (arrows) in the hair follicle of Piezo2WT (b) and Piezo2CKO dorsal skin (e and f). c, g, Representative image of immunostaining of S100 (green) and Nefh (red) in Meissner’s corpuscles in Piezo2WT (c) and Piezo2CKO (g) glabrous skin. Bg, bulge of the hair follicle; Der, dermis; Epi, epidermis; HS, hair shaft. Scale bars, 20 μm.

Extended Data Figure 5 Physiological properties of nociceptors are unaffected in Piezo2CKO mice.

a, No change in conduction velocities of Aβ-, Aδ- and C-fibre afferents in Piezo2CKO compared to Piezo2WT (Mann–Whitney test). b, Proportions of receptor types encountered among Aβ, Aδ and C fibres are shown. A-M, Aδ mechanonociceptor; C-M, C mechanonociceptor; C-MH, C mechano/heat receptor, responding both to noxious heat and mechanical stimuli; RAM, rapidly adapting mechanoreceptor; SAM, slowly adapting mechanoreceptor. c, Stimulus response properties of Aδ mechanonociceptors recorded in Piezo2CKO compared to Piezo2WT were not significantly different. d, D-hair receptors recorded from Piezo2CKO displayed stimulus response properties that were indistinguishable from control afferents. e, The stimulus response properties of C fibres in Piezo2CKO were not significantly different from C fibres recorded in control Piezo2WT mice. Data are presented as mean ± s.e.m., repeated measures ANOVA analysis for ce.

Extended Data Figure 6 Development of the novel two-choice mechanosensory assay.

a, Schematic of instrument construction and image of instrument from above. b, Schematic of the instrument from below, with the top cover removed, and photo of tactile transducers underneath the platform. c, Avoidance behaviour of C57BL/6J mice to the mechanically active side. Error bars represent s.e.m., n = 12 mice, 6 males and 6 females. **P < 0.005, ***P < 0.0001, Mann–Whitney non-parametric analysis.

Extended Data Figure 7 Piezo2CKO mice do not show deficits in noxious mechanical or thermal stimuli or in inflammatory pain responses.

a, Threshold for withdrawal response to a ramping protocol of von Frey stimulation from low force to high in Piezo2WT (n = 9) and Piezo2CKO (n = 7) mice. b, Time to response (latency) to application of a 500 g tail clip to the base of the tail in Piezo2WT (n = 6) and Piezo2CKO (n = 7) mice. c, Threshold for response to a Randall–Selitto pinching stimulus to the hind paw in Piezo2WT (n = 5) and Piezo2CKO (n = 7) mice. d, Time to withdrawal of hind paw in response to an infrared light heat source (Hargreaves assay) in Piezo2WT (n = 13) and Piezo2CKO (n = 9) mice. e, Ramping von Frey protocol in baseline (before CFA injection) and 24 h post CFA injection in Piezo2WT (n = 11) and Piezo2CKO (n = 9) mice. f, Ramping von Frey protocol in baseline (before bradykinin injection) and at time points 5, 15 and 30 min post injection in Piezo2WT (n = 6) and Piezo2CKO (n = 6) mice. Error bars represent s.e.m., all experiments performed with at least two separate cohorts of both male and female mice, **P < 0.005, ***P < 0.0001, Mann–Whitney non-parametric analysis.

Extended Data Figure 8 Piezo1 expression and function in DRGs.

a, In situ hybridization expression analysis of Piezo1 in DRG neurons relative to Piezo2. Robust expression of Piezo2 but not Piezo1 is observed, and this agrees with previously reported results using qPCR3. b, siRNA for Piezo1 in cultured DRG neurons does not affect the number of mechanically sensitive neurons or the ratio of rapidly, intermediately or slowly adapting currents (number of recorded neurons in each category indicated on top of bar graphs). Data from three independent preparations, not significant by Student’s t-test. Scale bar, 100 μm. KD, knockdown; scr, scrambled.

Extended Data Table 1 Electrical properties of Piezo2WT and Piezo2CKO cultured DRG neurons
Extended Data Table 2 Properties of single fibres recorded from Piezo2WT and Piezo2CKO using the saphenous nerve ex vivo skin nerve preparation

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ranade, S., Woo, SH., Dubin, A. et al. Piezo2 is the major transducer of mechanical forces for touch sensation in mice. Nature 516, 121–125 (2014). https://doi.org/10.1038/nature13980

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13980

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing