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:

The lysosomal potassium channel TMEM175 adopts a novel tetrameric architecture

Abstract

TMEM175 is a lysosomal K+ channel that is important for maintaining the membrane potential and pH stability in lysosomes1. It contains two homologous copies of a six-transmembrane-helix (6-TM) domain, which has no sequence homology to the canonical tetrameric K+ channels and lacks the TVGYG selectivity filter motif found in these channels2,3,4. The prokaryotic TMEM175 channel, which is present in a subset of bacteria and archaea, contains only a single 6-TM domain and functions as a tetramer. Here, we present the crystal structure of a prokaryotic TMEM175 channel from Chamaesiphon minutus, CmTMEM175, the architecture of which represents a completely different fold from that of canonical K+ channels. All six transmembrane helices of CmTMEM175 are tightly packed within each subunit without undergoing domain swapping. The highly conserved TM1 helix acts as the pore-lining inner helix, creating an hourglass-shaped ion permeation pathway in the channel tetramer. Three layers of hydrophobic residues on the carboxy-terminal half of the TM1 helices form a bottleneck along the ion conduction pathway and serve as the selectivity filter of the channel. Mutagenesis analysis suggests that the first layer of the highly conserved isoleucine residues in the filter is primarily responsible for channel selectivity. Thus, the structure of CmTMEM175 represents a novel architecture of a tetrameric cation channel whose ion selectivity mechanism appears to be distinct from that of the classical K+ channel family.

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

Figure 1: 86Rb flux assay of CmTMEM175.
Figure 2: Overall structure of CmTMEM175.
Figure 3: CmTMEM175 ion conduction pore.
Figure 4: Selectivity analysis of hTMEM175 and CmTMEM175.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

References

  1. Cang, C., Aranda, K., Seo, Y. J., Gasnier, B. & Ren, D. TMEM175 is an organelle K+ channel regulating lysosomal function. Cell 162, 1101–1112 (2015)

    Article  CAS  PubMed  Google Scholar 

  2. Papazian, D. M., Schwarz, T. L., Tempel, B. L., Jan, Y. N. & Jan, L. Y. Cloning of genomic and complementary DNA from Shaker, a putative potassium channel gene from Drosophila. Science 237, 749–753 (1987)

    Article  CAS  ADS  PubMed  Google Scholar 

  3. Heginbotham, L., Lu, Z., Abramson, T. & MacKinnon, R. Mutations in the K+ channel signature sequence. Biophys. J. 66, 1061–1067 (1994)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhou, Y., Morais-Cabral, J. H., Kaufman, A. & MacKinnon, R. Chemistry of ion coordination and hydration revealed by a K+ channel–Fab complex at 2.0 A resolution. Nature 414, 43–48 (2001)

    Article  CAS  ADS  PubMed  Google Scholar 

  5. Xu, H. & Ren, D. Lysosomal physiology. Annu. Rev. Physiol. 77, 57–80 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Luzio, J. P., Pryor, P. R. & Bright, N. A. Lysosomes: fusion and function. Nat. Rev. Mol. Cell Biol. 8, 622–632 (2007)

    Article  CAS  PubMed  Google Scholar 

  7. Kolter, T. & Sandhoff, K. Principles of lysosomal membrane digestion: stimulation of sphingolipid degradation by sphingolipid activator proteins and anionic lysosomal lipids. Annu. Rev. Cell Dev. Biol. 21, 81–103 (2005)

    Article  CAS  PubMed  Google Scholar 

  8. Vitner, E. B., Platt, F. M. & Futerman, A. H. Common and uncommon pathogenic cascades in lysosomal storage diseases. J. Biol. Chem. 285, 20423–20427 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Schulze, H. & Sandhoff, K. Lysosomal lipid storage diseases. Cold Spring Harb. Perspect. Biol. 3, a004804 (2011)

    Article  PubMed  PubMed Central  Google Scholar 

  10. Parkinson-Lawrence, E. J. et al. Lysosomal storage disease: revealing lysosomal function and physiology. Physiology (Bethesda) 25, 102–115 (2010)

  11. Mindell, J. A. Lysosomal acidification mechanisms. Annu. Rev. Physiol. 74, 69–86 (2012)

    Article  CAS  PubMed  Google Scholar 

  12. Xiong, J. & Zhu, M. X. Regulation of lysosomal ion homeostasis by channels and transporters. Sci. China Life Sci. 59, 777–791 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Jinn, S. et al. TMEM175 deficiency impairs lysosomal and mitochondrial function and increases α-synuclein aggregation. Proc. Natl Acad. Sci. USA 114, 2389–2394 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Nalls, M. A. et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat. Genet. 46, 989–993 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Heginbotham, L., Kolmakova-Partensky, L. & Miller, C. Functional reconstitution of a prokaryotic K+ channel. J. Gen. Physiol. 111, 741–749 (1998)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Long, S. B., Campbell, E. B. & Mackinnon, R. Crystal structure of a mammalian voltage-dependent Shaker family K+ channel. Science 309, 897–903 (2005)

    Article  CAS  ADS  PubMed  Google Scholar 

  17. Long, S. B., Tao, X., Campbell, E. B. & MacKinnon, R. Atomic structure of a voltage-dependent K+ channel in a lipid membrane-like environment. Nature 450, 376–382 (2007)

    Article  CAS  ADS  PubMed  Google Scholar 

  18. Holm, L. & Rosenström, P. Dali server: conservation mapping in 3D. Nucleic Acids Res. 38, W545–549 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Doyle, D. A. et al. The structure of the potassium channel: molecular basis of K+ conduction and selectivity. Science 280, 69–77 (1998)

    Article  CAS  ADS  PubMed  Google Scholar 

  20. Shi, N., Ye, S., Alam, A., Chen, L. & Jiang, Y. Atomic structure of a Na+- and K+-conducting channel. Nature 440, 570–574 (2006)

    Article  CAS  ADS  PubMed  Google Scholar 

  21. Aryal, P., Sansom, M. S. & Tucker, S. J. Hydrophobic gating in ion channels. J. Mol. Biol. 427, 121–130 (2015)

    Article  CAS  PubMed  Google Scholar 

  22. Yang, T. et al. Structure and selectivity in bestrophin ion channels. Science 346, 355–359 (2014)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  23. Kane Dickson, V., Pedi, L. & Long, S. B. Structure and insights into the function of a Ca2+-activated Cl channel. Nature 516, 213–218 (2014)

    Article  ADS  PubMed  Google Scholar 

  24. Hartzell, H. C., Qu, Z., Yu, K., Xiao, Q. & Chien, L. T. Molecular physiology of bestrophins: multifunctional membrane proteins linked to best disease and other retinopathies. Physiol. Rev. 88, 639–672 (2008)

    Article  CAS  PubMed  Google Scholar 

  25. Vaisey, G., Miller, A. N. & Long, S. B. Distinct regions that control ion selectivity and calcium-dependent activation in the bestrophin ion channel. Proc. Natl Acad. Sci. USA 113, E7399–E7408 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li, H., Francisco, J. S. & Zeng, X. C. Unraveling the mechanism of selective ion transport in hydrophobic subnanometer channels. Proc. Natl Acad. Sci. USA 112, 10851–10856 (2015)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  27. Zhou, X. et al. Self-assembling subnanometer pores with unusual mass-transport properties. Nat. Commun. 3, 949 (2012)

    Article  ADS  PubMed  Google Scholar 

  28. Carrillo-Tripp, M., Saint-Martin, H. & Ortega-Blake, I. Minimalist molecular model for nanopore selectivity. Phys. Rev. Lett. 93, 168104 (2004)

    Article  ADS  PubMed  Google Scholar 

  29. Shao, Q. et al. Anomalous hydration shell order of Na+ and K+ inside carbon nanotubes. Nano Lett. 9, 989–994 (2009)

    Article  CAS  ADS  PubMed  Google Scholar 

  30. Otwinowski, Z. & Minor, W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276, 307–326 (1997)

    Article  CAS  PubMed  Google Scholar 

  31. Strong, M. et al. Toward the structural genomics of complexes: crystal structure of a PE/PPE protein complex from Mycobacterium tuberculosis. Proc. Natl Acad. Sci. USA 103, 8060–8065 (2006)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  32. Schneider, T. R. & Sheldrick, G. M. Substructure solution with SHELXD. Acta Crystallogr. D Biol. Crystallogr. 58, 1772–1779 (2002)

    Article  PubMed  Google Scholar 

  33. Vonrhein, C., Blanc, E., Roversi, P. & Bricogne, G. Automated structure solution with autoSHARP. Methods Mol. Biol. 364, 215–230 (2007)

    CAS  PubMed  Google Scholar 

  34. Abrahams, J. P. & Leslie, A. G. W. Methods used in the structure determination of bovine mitochondrial F1 ATPase. Acta Crystallogr. D Biol. Crystallogr. 52, 30–42 (1996)

    Article  CAS  PubMed  Google Scholar 

  35. Terwilliger, T. SOLVE and RESOLVE: automated structure solution, density modification and model building. J. Synchrotron Radiat. 11, 49–52 (2004)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Laskowski, R. A., MacArthur, M. W., Moss, D. S. & Thornton, J. M. PROCHECK: a program to check the stereochemical quality of protein structures. J. Appl. Crystallogr. 26, 283–291 (1993)

    Article  CAS  Google Scholar 

  39. Smart, O. S., Goodfellow, J. M. & Wallace, B. A. The pore dimensions of gramicidin A. Biophys. J. 65, 2455–2460 (1993)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Baker, N. A., Sept, D., Joseph, S., Holst, M. J. & McCammon, J. A. Electrostatics of nanosystems: application to microtubules and the ribosome. Proc. Natl Acad. Sci. USA 98, 10037–10041 (2001)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  41. The PyMOL Molecular Graphics System Version 1.7.4 (Schrödinger, LLC)

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

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank N. Nguyen for manuscript preparation. The experimental results reported in this article derive from work performed at Argonne National Laboratory, Structural Biology Center (19ID) and GM/CA (23ID) at the Advanced Photon Source, and from work performed at the Berkeley Center for Structural Biology at the Advanced Light Source (ALS). Argonne is operated by UChicago Argonne, LLC, for the US Department of Energy, Office of Biological and Environmental Research under contract DE-AC02-06CH11357. The Berkeley Center for Structural Biology is supported in part by the National Institutes of Health, National Institute of General Medical Sciences, and the Howard Hughes Medical Institute. The Advanced Light Source is supported by the Director, Office of Science, Office of Basic Energy Sciences, of the US Department of Energy under Contract No. DE-AC02-05CH11231. This work was supported in part by the Howard Hughes Medical Institute and by grants from the National Institutes of Health (GM079179 to Y.J.; NS055293 and NS074257 to D.R.) and the Welch Foundation (Grant I-1578 to Y.J.).

Author information

Authors and Affiliations

Authors

Contributions

C.L. and J.G. performed the structure determination; W.Z., C.C. and D.R. performed electrophysiology; S.K and J.S. participated in sample preparation; C.L., J.G., W.Z. and Y.J. designed the research, analysed data, and prepared the manuscript.

Corresponding author

Correspondence to Youxing Jiang.

Ethics declarations

Competing interests

The authors declare no competing financial 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 Figure 1 Sequence comparison between prokaryotic and eukaryotic TMEM175 channels.

a, Topologies of prokaryotic (left) and eukaryotic (right) TMEM175 channel subunits. b, Sequence alignment of prokaryotic TMEM175 proteins and the first 6-TM domains of eukaryotic TMEM175 proteins. c, Sequence alignment of prokaryotic TMEM175 proteins and the second 6-TM domains of eukaryotic TMEM175 proteins. Secondary structure assignments are based on the CmTMEM175 structure. Asterisks mark the three hydrophobic filter residues. Blue triangles mark the RxxxFSD motif and the residues participating in the inter- and intra-subunit interactions with the motif.

Extended Data Figure 2 Biochemical analysis of bacterial TMEM175 channels.

a, Gel filtration profiles of three purified bacTMEM175 channels on Superdex-200 (10/30 GL column) in 20 mM HEPES, pH 7.4, 200 mM KCl and 3 mM DM. All purified bacTMEM175 channels eluted as a monodispersed peak at a position much larger than a monomer, indicating oligomerization. Arrows indicate the elution peaks of three standard proteins (29, 66 and 200 KDa) on the same column. b, Cross-linking reaction of purified bacTMEM175 channels from Streptomyces collinus (left gel) and Chryseobacterium sp. (right gel) with the cross-linking reagents DSS and DSG. Samples were analysed by SDS–PAGE and detected by coomassie blue staining. The purified proteins migrate as multiple bands on SDS–PAGE corresponding to the sizes of monomer to tetramer. The cross-linking reaction promotes the formation of a cross-linked tetramer, demonstrating that these bacterial channels form tetramers in solution. For gel source data, see Supplementary Fig. 1.

Extended Data Figure 3 Electron density maps of CmTMEM175.

a, 2Fo−Fc electron density map of one subunit (contoured at 1.5σ). b, 2Fo−Fc electron density map of the filter region (contoured at 1.5σ). The front and back subunits have been removed for clarity. The side chains of the three layers of hydrophobic residues are coloured in magenta.

Extended Data Figure 4 Structural comparison between K+ channels.

a, CmTMEM175; b, Shaker-like K+ channel (Kv1.2-2.1 chimaera, PDB code: 2R9R). Each subunit is individually coloured. Both structures are viewed from the extracellular side.

Extended Data Figure 5 Structural comparison of the ion conduction pathways.

Ion conduction pathways are shaded grey. a, CmTMEM175; b, chicken bestrophin-1 (PDB code: 4RDQ); c, bacterial bestrophin from Klebsiella pneumoniae (KpBest, PDB code: 4WD8). Insets show zoomed-in views of the narrow filters.

Extended Data Figure 6 Ion selectivity and pharmacological properties of human TMEM175.

a, Partial sequence alignment of TM1 helices from hTMEM175 and CmTMEM175; the three layers of hydrophobic residues are boxed and the RxxxFSD motif is shaded red. b, IV curve from a control cell. The pipette and bath solutions contained 150 mM Cs+ and 150 mM Na+, respectively. c, Extracellular Zn2+ and 4-AP blockade of human TMEM175. Currents were recorded using whole-cell patches with 150 mM extracellular Na+ (bath) and 150 mM intracellular Cs+ (pipette). d, Intracellular Zn2+ and 4-AP blockade of human TMEM175. Currents were recorded using inside-out patches with 150 mM intracellular Na+ (bath) and 150 mM extracellular Cs+ (pipette). Recordings shown in c and d indicate that human TMEM175 is sensitive to Zn2+ or 4-AP block from both sides. e, IV curves of wild-type human TMEM175. Currents were recorded using whole-cell patches in bi-ionic conditions. The pipette solution contained 150 mM Cs+ and the bath solution contained 150 mM X+ (X = NMDG, Li, Na, K or Rb). f, IV curves of the I46N/I271N (at layer 1) double mutant of human TMEM175 in bi-ionic conditions. Currents were recorded using the same conditions as e. g, Summary of reversal potentials of hTMEM175 and its mutants and the calculated relative permeability between Cs+ and K+ or Na+; shown are mean ± s.e.m. of ≥5 measurements. h, Summary of reversal potentials of human TMEM175 and I46N/I271N mutant with various monovalent cations in the bath solutions and the calculated relative permeability of these ions in comparison to Cs+.

Extended Data Table 1 Data collection and structure refinement statistics

Supplementary information

Supplementary Information

This file contains the uncropped scan with size marker indications.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, C., Guo, J., Zeng, W. et al. The lysosomal potassium channel TMEM175 adopts a novel tetrameric architecture. Nature 547, 472–475 (2017). https://doi.org/10.1038/nature23269

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature23269

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