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:

Architecture of the TRPM2 channel and its activation mechanism by ADP-ribose and calcium

Abstract

Transient receptor potential melastatin 2 (TRPM2) is a calcium-permeable, non-selective cation channel that has an essential role in diverse physiological processes such as core body temperature regulation, immune response and apoptosis1,2,3,4. TRPM2 is polymodal and can be activated by a wide range of stimuli1,2,3,4,5,6,7, including temperature, oxidative stress and NAD+-related metabolites such as ADP-ribose (ADPR). Its activation results in both Ca2+ entry across the plasma membrane and Ca2+ release from lysosomes8, and has been linked to diseases such as ischaemia-reperfusion injury, bipolar disorder and Alzheimer’s disease9,10,11. Here we report the cryo-electron microscopy structures of the zebrafish TRPM2 in the apo resting (closed) state and in the ADPR/Ca2+-bound active (open) state, in which the characteristic NUDT9-H domains hang underneath the MHR1/2 domain. We identify an ADPR-binding site located in the bi-lobed structure of the MHR1/2 domain. Our results provide an insight into the mechanism of activation of the TRPM channel family and define a framework for the development of therapeutic agents to treat neurodegenerative diseases and temperature-related pathological conditions.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: The overall architecture.
Fig. 2: The ion-conducting pore.
Fig. 3: The ligand-sensing layer.
Fig. 4: Signal transduction through the linker layer MHR3/4.
Fig. 5: Signal transduction in the transmembrane domain from EDTA–TRPM2 to ADPR/Ca2+–TRPM2.
Fig. 6: Schematic of the activation mechanism of TRPM2.

Similar content being viewed by others

Data availability

The cryo-electron microscopy density map and coordinates of EDTA–TRPM2 and ADPR/Ca2+–TRPM2 have been deposited in the Electron Microscopy Data Bank (EMDB) under accession numbers EMD-8901 and EMD-7999 and in the Research Collaboratory for Structural Bioinformatics Protein Data Bank under accession codes 6DRK and 6DRJ. All other data relating to this study are available from the corresponding author upon reasonable request.

References

  1. Tan, C. H. & McNaughton, P. A. The TRPM2 ion channel is required for sensitivity to warmth. Nature 536, 460–463 (2016).

    Article  ADS  CAS  Google Scholar 

  2. Song, K. et al. The TRPM2 channel is a hypothalamic heat sensor that limits fever and can drive hypothermia. Science 353, 1393–1398 (2016).

    Article  ADS  CAS  Google Scholar 

  3. Knowles, H. et al. Transient Receptor Potential Melastatin 2 (TRPM2) ion channel is required for innate immunity against Listeria monocytogenes. Proc. Natl Acad. Sci. USA 108, 11578–11583 (2011).

    Article  ADS  CAS  Google Scholar 

  4. Hecquet, C. M. et al. Cooperative interaction of trp melastatin channel transient receptor potential (TRPM2) with its splice variant TRPM2 short variant is essential for endothelial cell apoptosis. Circ. Res. 114, 469–479 (2014).

    Article  CAS  Google Scholar 

  5. Kolisek, M., Beck, A., Fleig, A. & Penner, R. Cyclic ADP-ribose and hydrogen peroxide synergize with ADP-ribose in the activation of TRPM2 channels. Mol. Cell 18, 61–69 (2005).

    Article  CAS  Google Scholar 

  6. Beck, A., Kolisek, M., Bagley, L. A., Fleig, A. & Penner, R. Nicotinic acid adenine dinucleotide phosphate and cyclic ADP-ribose regulate TRPM2 channels in T lymphocytes. FASEB J. 20, 962–964 (2006).

    Article  CAS  Google Scholar 

  7. Togashi, K. et al. TRPM2 activation by cyclic ADP-ribose at body temperature is involved in insulin secretion. EMBO J. 25, 1804–1815 (2006).

    Article  CAS  Google Scholar 

  8. Lange, I. et al. TRPM2 functions as a lysosomal Ca2+-release channel in β cells. Sci. Signal. 2, ra23 (2009).

    Article  Google Scholar 

  9. Miller, B. A. et al. TRPM2 channels protect against cardiac ischemia-reperfusion injury: role of mitochondria. J. Biol. Chem. 289, 7615–7629 (2014).

    Article  CAS  Google Scholar 

  10. Xu, C. et al. Association of the putative susceptibility gene, transient receptor potential protein melastatin type 2, with bipolar disorder. Am. J. Med. Genet. B 141B, 36–43 (2006).

    Article  CAS  Google Scholar 

  11. Ostapchenko, V. G. et al. The transient receptor potential melastatin 2 (TRPM2) channel contributes to β-amyloid oligomer-related neurotoxicity and memory impairment. J. Neurosci. 35, 15157–15169 (2015).

    Article  CAS  Google Scholar 

  12. Perraud, A. L. et al. ADP-ribose gating of the calcium-permeable LTRPC2 channel revealed by Nudix motif homology. Nature 411, 595–599 (2001).

    Article  ADS  CAS  Google Scholar 

  13. Perraud, A. L. et al. Accumulation of free ADP-ribose from mitochondria mediates oxidative stress-induced gating of TRPM2 cation channels. J. Biol. Chem. 280, 6138–6148 (2005).

    Article  CAS  Google Scholar 

  14. Wehage, E. et al. Activation of the cation channel long transient receptor potential channel 2 (LTRPC2) by hydrogen peroxide. J. Biol. Chem. 277, 23150–23156 (2002).

    Article  CAS  Google Scholar 

  15. Kühn, F. J., Kühn, C., Winking, M., Hoffmann, D. C. & Lückhoff, A. ADP-ribose activates the TRPM2 channel from the sea anemone Nematostella vectensis independently of the NUDT9H domain. PLoS ONE 11, e0158060 (2016).

    Article  Google Scholar 

  16. Kühn, F. J. & Lückhoff, A. Sites of the NUDT9-H domain critical for ADP-ribose activation of the cation channel TRPM2. J. Biol. Chem. 279, 46431–46437 (2004).

    Article  Google Scholar 

  17. Yu, P. et al. Identification of the ADPR binding pocket in the NUDT9 homology domain of TRPM2. J. Gen. Physiol. 149, 219–235 (2017).

    Article  ADS  Google Scholar 

  18. Fliegert, R. et al. Ligand-induced activation of human TRPM2 requires the terminal ribose of ADPR and involves Arg1433 and Tyr1349. Biochem. J. 474, 2159–2175 (2017).

    Article  CAS  Google Scholar 

  19. Iordanov, I., Mihályi, C., Tóth, B. & Csanády, L. The proposed channel-enzyme transient receptor potential melastatin 2 does not possess ADP ribose hydrolase activity. eLife 5, e17600 (2016).

    Article  Google Scholar 

  20. Tóth, B., Iordanov, I. & Csanády, L. Putative chanzyme activity of TRPM2 cation channel is unrelated to pore gating. Proc. Natl Acad. Sci. USA 111, 16949–16954 (2014).

    Article  ADS  Google Scholar 

  21. Maruyama, Y. et al. Three-dimensional reconstruction using transmission electron microscopy reveals a swollen, bell-shaped structure of transient receptor potential melastatin type 2 cation channel. J. Biol. Chem. 282, 36961–36970 (2007).

    Article  CAS  Google Scholar 

  22. Mei, Z. Z., Mao, H. J. & Jiang, L. H. Conserved cysteine residues in the pore region are obligatory for human TRPM2 channel function. Am. J. Physiol. Cell Physiol. 291, C1022–C1028 (2006).

    Article  CAS  Google Scholar 

  23. Zhang, Z., Toth, B., Szollosi, A., Chen, J. & Csanady, L. Structure of a TRPM2 channel in complex with Ca2+ explains unique gating regulation. eLife 7, (2018).

  24. Winkler, P. A., Huang, Y., Sun, W., Du, J. & Lü, W. Electron cryo-microscopy structure of a human TRPM4 channel. Nature 552, 200–204 (2017).

    Article  ADS  CAS  Google Scholar 

  25. Liao, M., Cao, E., Julius, D. & Cheng, Y. Structure of the TRPV1 ion channel determined by electron cryo-microscopy. Nature 504, 107–112 (2013).

    Article  ADS  CAS  Google Scholar 

  26. Guo, J. et al. Structures of the calcium-activated, non-selective cation channel TRPM4. Nature 552, 205–209 (2017).

    Article  ADS  CAS  Google Scholar 

  27. Yin, Y. et al. Structure of the cold- and menthol-sensing ion channel TRPM8. Science 359, 237–241 (2018).

    Article  ADS  CAS  Google Scholar 

  28. Autzen, H. E. et al. Structure of the human TRPM4 ion channel in a lipid nanodisc. Science 359, 228–232 (2018).

    Article  ADS  CAS  Google Scholar 

  29. Grimm, C., Kraft, R., Sauerbruch, S., Schultz, G. & Harteneck, C. Molecular and functional characterization of the melastatin-related cation channel TRPM3. J. Biol. Chem. 278, 21493–21501 (2003).

    Article  CAS  Google Scholar 

  30. Nadler, M. J. et al. LTRPC7 is a Mg·ATP-regulated divalent cation channel required for cell viability. Nature 411, 590–595 (2001).

    Article  ADS  CAS  Google Scholar 

  31. Voets, T. et al. TRPM6 forms the Mg2+ influx channel involved in intestinal and renal Mg2+ absorption. J. Biol. Chem. 279, 19–25 (2004).

    Article  CAS  Google Scholar 

  32. Lambert, S. et al. Transient receptor potential melastatin 1 (TRPM1) is an ion-conducting plasma membrane channel inhibited by zinc ions. J. Biol. Chem. 286, 12221–12233 (2011).

    Article  CAS  Google Scholar 

  33. Xia, R. et al. Identification of pore residues engaged in determining divalent cationic permeation in transient receptor potential melastatin subtype channel 2. J. Biol. Chem. 283, 27426–27432 (2008).

    Article  CAS  Google Scholar 

  34. Kashio, M. et al. Redox signal-mediated sensitization of transient receptor potential melastatin 2 (TRPM2) to temperature affects macrophage functions. Proc. Natl Acad. Sci. USA 109, 6745–6750 (2012).

    Article  ADS  CAS  Google Scholar 

  35. Gregorio-Teruel, L. et al. The Integrity of the TRP domain is pivotal for correct TRPV1 channel gating. Biophys. J. 109, 529–541 (2015).

    Article  ADS  CAS  Google Scholar 

  36. Winking, M. et al. Importance of a conserved sequence motif in transmembrane segment S3 for the gating of human TRPM8 and TRPM2. PLoS ONE 7, e49877 (2012).

    Article  ADS  CAS  Google Scholar 

  37. Goehring, A. et al. Screening and large-scale expression of membrane proteins in mammalian cells for structural studies. Nat. Protoc. 9, 2574–2585 (2014).

    Article  CAS  Google Scholar 

  38. Kawate, T. & Gouaux, E. Fluorescence-detection size-exclusion chromatography for precrystallization screening of integral membrane proteins. Structure 14, 673–681 (2006).

    Article  CAS  Google Scholar 

  39. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  Google Scholar 

  40. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  Google Scholar 

  41. Zhang, K. Gctf: Real-time CTF determination and correction. J. Struct. Biol. 193, 1–12 (2016).

    Article  ADS  CAS  Google Scholar 

  42. Scheres, S. H. RELION: implementation of a Bayesian approach to cryo-EM structure determination. J. Struct. Biol. 180, 519–530 (2012).

    Article  CAS  Google Scholar 

  43. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    Article  CAS  Google Scholar 

  44. Grigorieff, N. Frealign: an exploratory tool for single-particle cryo-EM. Methods Enzymol. 579, 191–226 (2016).

    Article  CAS  Google Scholar 

  45. Heymann, J. B. Guidelines for using Bsoft for high resolution reconstruction and validation of biomolecular structures from electron micrographs. Protein Sci. 27, 159–171 (2018).

    Article  CAS  Google Scholar 

  46. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    Article  CAS  Google Scholar 

  47. Arnold, K., Bordoli, L., Kopp, J. & Schwede, T. The SWISS-MODEL workspace: a web-based environment for protein structure homology modelling. Bioinformatics 22, 195–201 (2006).

    Article  CAS  Google Scholar 

  48. Shen, B. W., Perraud, A. L., Scharenberg, A. & Stoddard, B. L. The crystal structure and mutational analysis of human NUDT9. J. Mol. Biol. 332, 385–398 (2003).

    Article  CAS  Google Scholar 

  49. Trabuco, L. G., Villa, E., Schreiner, E., Harrison, C. B. & Schulten, K. Molecular dynamics flexible fitting: a practical guide to combine cryo-electron microscopy and X-ray crystallography. Methods 49, 174–180 (2009).

    Article  CAS  Google Scholar 

  50. Afonine, P. V. et al. Towards automated crystallographic structure refinement with phenix.refine. Acta Crystallogr. D 68, 352–367 (2012).

    Article  CAS  Google Scholar 

  51. Chen, V. B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D 66, 12–21 (2010).

    Article  CAS  Google Scholar 

  52. Barad, B. A. et al. EMRinger: side chain-directed model and map validation for 3D cryo-electron microscopy. Nat. Methods 12, 943–946 (2015).

    Article  CAS  Google Scholar 

  53. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  Google Scholar 

  54. Grant, T., Rohou, A. & Grigorieff, N. cisTEM, user-friendly software for single-particle image processing. eLife 7, e35383 (2018).

    Article  Google Scholar 

  55. Sievers, F. et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol. Syst. Biol. 7, 539 (2011).

    Article  Google Scholar 

  56. Drozdetskiy, A., Cole, C., Procter, J. & Barton, G. J. JPred4: a protein secondary structure prediction server. Nucleic Acids Res. 43, W389–W394 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank G. Zhao and X. Meng for support with data collection at the David Van Andel Advanced Cryo-Electron Microscopy Suite, the HPC team in the Van Andel Research Institute (VARI) for computational support, C. Xu for help with SerialEM, and D. Nadziejka for technical editing. This work was supported by internal VARI funding.

Reviewer information

Nature thanks A. H. Guse, A. Ward and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

J.D. and W.L. initiated the project. Y.H. and P.A.W. purified TRPM2; Y.H. and W.S. performed electrophysiological experiments; and Y.H., J.D. and W.L. performed cryo-electron microscopy data collection, processing, analysis and wrote the manuscript. All authors contributed to the preparation of the manuscript.

Corresponding authors

Correspondence to Wei Lü or Juan Du.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 The cryo-electron microscopy data processing workflow, using the data of EDTA–TRPM2 as an example.

Particles were auto-picked using Gautomatch and visually checked in RELION. After removing false positives, particles were subjected to two rounds of 2D classification in RELION. The contrast transfer function values of individual particles from selected 2D class averages were estimated using Gctf. For 3D classification in RELION, a reference model was generated using CryoSparc. Initial 3D refinement was carried out using RELION. Particles were further refined using cisTEM54.

Extended Data Fig. 2 Cryo-electron microscopy analysis of full-length DrTRPM2 in the presence of EDTA and ADPR/Ca2+.

a, e, Representative electron micrograph of EDTA–TRPM2 (a) and ADPR/Ca2+–TRPM2 (e). b, f, Selected 2D class averages of the electron micrographs of EDTA–TRPM2 (b) and ADPR/Ca2+–TRPM2 (f). c, g, The gold-standard Fourier shell correlation curves for the electron microscopy maps of EDTA–TRPM2 (c) and ADPR/Ca2+–TRPM2 (g) are shown in black, and the Fourier shell correlation curves between the atomic model and the final electron microscopy maps are shown in blue. d, h, Angular distribution of particles used for the refinement of EDTA–TRPM2 (d) and ADPR/Ca2+–TRPM2 (h).

Extended Data Fig. 3 Representative densities of the reconstruction of EDTA–TRPM2 and ADPR/Ca2+–TRPM2.

a, b, Local resolution estimation of the structure of EDTA–TRPM2. The map is coloured according to local resolution estimation. The unsharpened reconstructions are shown as transparent envelopes. c, Representative densities of EDTA–TRPM2. d, e, Local resolution estimation of the structure of ADPR/Ca2+–TRPM2. f, Representative densities of ADPR/Ca2+–TRPM2.

Extended Data Fig. 4 Overall architecture of TRPM2.

a, Domain organization of TRPM2. Dashed lines and cylinders denote regions that have not been modelled. b, c, Cartoon representation of one subunit of the EDTA–TRPM2 structure. d, e, Cartoon representation of one subunit of the ADPR/Ca2+–TRPM2 structure. ADPR and Ca2+ are shown as spheres.

Extended Data Fig. 5 The NUDT9-H domain.

a, Sequence alignment of zebrafish (Dr), starlet sea anemone (Nv) and human (Hs) TRPM2 NUDT9-H domains with human NUDT9 using Clustal Omega55. b, c, Superimposition of the zebrafish ADPR/Ca2+–TRPM2 NUDT9-H domain (red) with human NUDT9 (green, PDB ID: 1Q33). Cap and core regions are indicated. d, e, Superimposition of the NUDT9-H domains of the zebrafish ADPR/Ca2+–TRPM2 structure (red) and the EDTA–TRPM2 structure (blue).

Extended Data Fig. 6 Comparison of NvTRPM2 with DrTRPM2 (EDTA–TRPM2 and ADPR/Ca2+–TRPM2) and HsTRPM4, and comparison of the gate and selectivity filter of EDTA–TRPM2 with those of HsTRPM4 (PDB: 5WP6).

ac, Superimposition of EDTA–TRPM2 (a, blue, r.m.s.d = 50 Å, overall, main chain atoms only), ADPR/Ca2+–TRPM2 (b, red, r.m.s.d = 50 Å, overall, main chain atoms only) and HsTRPM4 (c, green, r.m.s.d = 47.5 Å, overall, main chain atoms only) with NvTRPM2 (yellow). The NUDT9-H domain is completely invisible in NvTRPM2. df, Superimposition of the MHR1/2 domains of EDTA–TRPM2 (d, blue), ADPR/Ca2+–TRPM2 (e, red) and HsTRPM4 (f, green) with NvTRPM2 (yellow). gi, Superimposition of the transmembrane domains of EDTA–TRPM2 (g, blue), ADPR/Ca2+–TRPM2 (h, red) and HsTRPM4 (i, green) with NvTRPM2 (yellow). The transmembrane domain of NvTRPM2 is distinct from EDTA–TRPM2 and ADPR/Ca2+–TRPM2 but very similar to that of HsTRPM4. j, k, Comparison of the gates of TRPM2 (blue) and TRPM4 (green) viewed from the intracellular side of the membrane (j), or viewed parallel to the membrane (k). Only two subunits are shown in (k) for clarity. l, m, Comparison of the selectivity filters of DrTRPM2 and HsTRPM4 viewed from the extracellular side of the membrane (l), or viewed parallel to the membrane (m). Only two subunits are shown in m for clarity. The superimposition was performed by aligning the P loop and S6 (residues 958–1050 in HsTRPM4 and residues 978–1069 in DrTRPM2). The Cα atoms of the residues in the selectivity filter and gate are shown as spheres.

Extended Data Fig. 7 Secondary structure arrangement of DrTRPM2 and sequence alignment of TRPM family channels.

The secondary structure prediction of DrTRPM2 was performed using the JPred online server56. The sequences (TRPM2 from zebrafish; TRPM2, TRPM4, TRPM5 and TRPM8 from human; TRPM2 from starlet sea anemone) were aligned using Clustal Omega55. Residues that are involved in ADPR binding and calcium binding are marked with red and black triangles, respectively. The cap and core regions of the NUDT9-H domain are indicated.

Extended Data Fig. 8 Electrophysiological experiments.

a, b, Alanine mutations were introduced within the ADPR-binding pocket and showed expression levels comparable to that of wild-type TRPM2, observed from both transfected cells (a) and fluorescence-detection size-exclusion chromatography (b). c, d, Electrophysiological experiments were carried out to measure the amplitude of agonist-induced current in inside-out patches pulled from HEK293 cells, with c showing the representative current and d showing the statistics of current amplitude and cell numbers. At +60 mV, robust current (4.87 ± 0.55 nA, n = 9 cells) could be detected when applying 0.1 mM ADPR and 1 mM Ca2+ onto inside-out patches expressing wild-type TRPM2. Single mutations (K154A, Y271A, E274A, R278A and R334A) each show robust channel activation (n = 5 cells, 8 cells, 8 cells, 7 cells and 8 cells for the corresponding mutants) and—other than K154A receptors, which did not show a markedly reduced current amplitude—mean current amplitudes for the mutated receptors were around two- to threefold smaller compared to the wild type. Introducing double mutations R278A/R334A (n = 7 cells) to the receptor nearly abolished ADPR/Ca2+-induced current. Deletion of the NUDT9-H domain (∆NUDT9-H) (n = 6 cells) also nearly abolished the ADPR/Ca2+-induced current. Data are shown as mean ± s.e.m.

Extended Data Fig. 9 Putative calcium-binding site.

a, Densities of the putative Ca2+-binding site and adjacent residues. b, Comparison of the Ca2+-binding site in DrTRPM2 (red) and HsTRPM4 (white). Residues coordinating the Ca2+ ion are indicated, with residues of HsTRPM4 shown in parentheses. c, Sequence alignment of the putative Ca2+-binding site within the TRPM family. Residues coordinating the Ca2+ ion are marked with a black triangle.

Extended Data Table 1 Statistics of 3D reconstruction and model refinement

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Huang, Y., Winkler, P.A., Sun, W. et al. Architecture of the TRPM2 channel and its activation mechanism by ADP-ribose and calcium. Nature 562, 145–149 (2018). https://doi.org/10.1038/s41586-018-0558-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0558-4

Keywords

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