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.

  • Article
  • Published:

Structure of a CLC chloride ion channel by cryo-electron microscopy

Abstract

CLC proteins transport chloride (Cl) ions across cellular membranes to regulate muscle excitability, electrolyte movement across epithelia, and acidification of intracellular organelles. Some CLC proteins are channels that conduct Cl ions passively, whereas others are secondary active transporters that exchange two Cl ions for one H+. The structural basis underlying these distinctive transport mechanisms is puzzling because CLC channels and transporters are expected to share the same architecture on the basis of sequence homology. Here we determined the structure of a bovine CLC channel (CLC-K) using cryo-electron microscopy. A conserved loop in the Cl transport pathway shows a structure markedly different from that of CLC transporters. Consequently, the cytosolic constriction for Cl passage is widened in CLC-K such that the kinetic barrier previously postulated for Cl/H+ transporter function would be reduced. Thus, reduction of a kinetic barrier in CLC channels enables fast flow of Cl down its electrochemical gradient.

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: Functional and cryo-EM analysis of a bovine CLC-K channel.
Figure 2: Flexibility of the CLC-K dimer arrangement.
Figure 3: Architecture of the CLC-K channel.
Figure 4: Cl ion transport pathway.
Figure 5: Cl ion accessibility in the pore region.
Figure 6: Models for the mechanisms of ion transport by CLC.

Similar content being viewed by others

Accession codes

Primary accessions

Electron Microscopy Data Bank

Protein Data Bank

References

  1. Jentsch, T. J. Discovery of CLC transport proteins: cloning, structure, function and pathophysiology. J. Physiol. 593, 4091–4109 (2015)

    Article  CAS  Google Scholar 

  2. Maduke, M., Miller, C. & Mindell, J. A. A decade of CLC chloride channels: structure, mechanism, and many unsettled questions. Annu. Rev. Biophys. Biomol. Struct. 29, 411–438 (2000)

    Article  CAS  Google Scholar 

  3. Miller, C. ClC chloride channels viewed through a transporter lens. Nature 440, 484–489 (2006)

    Article  ADS  CAS  Google Scholar 

  4. Koch, M. C. et al. The skeletal muscle chloride channel in dominant and recessive human myotonia. Science 257, 797–800 (1992)

    Article  ADS  CAS  Google Scholar 

  5. Simon, D. B. et al. Mutations in the chloride channel gene, CLCNKB, cause Bartter’s syndrome type III. Nature Genet. 17, 171–178 (1997)

    Article  CAS  Google Scholar 

  6. Fisher, S. E. et al. Isolation and partial characterization of a chloride channel gene which is expressed in kidney and is a candidate for Dent’s disease (an X-linked hereditary nephrolithiasis). Hum. Mol. Genet. 3, 2053–2059 (1994)

    CAS  PubMed  Google Scholar 

  7. Kornak, U. et al. Loss of the ClC-7 chloride channel leads to osteopetrosis in mice and man. Cell 104, 205–215 (2001)

    Article  CAS  Google Scholar 

  8. Miller, C. & White, M. M. Dimeric structure of single chloride channels from Torpedo electroplax. Proc. Natl Acad. Sci. USA 81, 2772–2775 (1984)

    Article  ADS  CAS  Google Scholar 

  9. Jentsch, T. J., Steinmeyer, K. & Schwarz, G. Primary structure of Torpedo marmorata chloride channel isolated by expression cloning in Xenopus oocytes. Nature 348, 510–514 (1990)

    Article  ADS  CAS  Google Scholar 

  10. Accardi, A. & Miller, C. Secondary active transport mediated by a prokaryotic homologue of ClC Cl channels. Nature 427, 803–807 (2004)

    Article  ADS  CAS  Google Scholar 

  11. Scheel, O., Zdebik, A. A., Lourdel, S. & Jentsch, T. J. Voltage-dependent electrogenic chloride/proton exchange by endosomal CLC proteins. Nature 436, 424–427 (2005)

    Article  ADS  CAS  Google Scholar 

  12. Picollo, A. & Pusch, M. Chloride/proton antiporter activity of mammalian CLC proteins ClC-4 and ClC-5. Nature 436, 420–423 (2005)

    Article  ADS  CAS  Google Scholar 

  13. Walden, M. et al. Uncoupling and turnover in a Cl/H+ exchange transporter. J. Gen. Physiol. 129, 317–329 (2007)

    Article  CAS  Google Scholar 

  14. Jayaram, H., Accardi, A., Wu, F., Williams, C. & Miller, C. Ion permeation through a Cl-selective channel designed from a CLC Cl/H+ exchanger. Proc. Natl Acad. Sci. USA 105, 11194–11199 (2008)

    Article  ADS  CAS  Google Scholar 

  15. Ludewig, U., Pusch, M. & Jentsch, T. J. Two physically distinct pores in the dimeric ClC-0 chloride channel. Nature 383, 340–343 (1996)

    Article  ADS  CAS  Google Scholar 

  16. Dutzler, R., Campbell, E. B., Cadene, M., Chait, B. T. & MacKinnon, R. X-ray structure of a ClC chloride channel at 3.0 Å reveals the molecular basis of anion selectivity. Nature 415, 287–294 (2002)

    Article  ADS  CAS  Google Scholar 

  17. Dutzler, R., Campbell, E. B. & MacKinnon, R. Gating the selectivity filter in ClC chloride channels. Science 300, 108–112 (2003)

    Article  ADS  CAS  Google Scholar 

  18. Feng, L., Campbell, E. B., Hsiung, Y. & MacKinnon, R. Structure of a eukaryotic CLC transporter defines an intermediate state in the transport cycle. Science 330, 635–641 (2010)

    Article  ADS  CAS  Google Scholar 

  19. Adachi, S. et al. Two isoforms of a chloride channel predominantly expressed in thick ascending limb of Henle’s loop and collecting ducts of rat kidney. J. Biol. Chem. 269, 17677–17683 (1994)

    CAS  PubMed  Google Scholar 

  20. Kieferle, S., Fong, P., Bens, M., Vandewalle, A. & Jentsch, T. J. Two highly homologous members of the ClC chloride channel family in both rat and human kidney. Proc. Natl Acad. Sci. USA 91, 6943–6947 (1994)

    Article  ADS  CAS  Google Scholar 

  21. Denton, J. S., Pao, A. C. & Maduke, M. Novel diuretic targets. Am. J. Physiol. Renal Physiol. 305, F931–F942 (2013)

    Article  CAS  Google Scholar 

  22. Estévez, R. et al. Barttin is a Cl channel β-subunit crucial for renal Cl reabsorption and inner ear K+ secretion. Nature 414, 558–561 (2001)

    Article  ADS  Google Scholar 

  23. Scholl, U. et al. Barttin modulates trafficking and function of ClC-K channels. Proc. Natl Acad. Sci. USA 103, 11411–11416 (2006)

    Article  ADS  CAS  Google Scholar 

  24. Waldegger, S. & Jentsch, T. J. Functional and structural analysis of ClC-K chloride channels involved in renal disease. J. Biol. Chem. 275, 24527–24533 (2000)

    Article  CAS  Google Scholar 

  25. L’Hoste, S. et al. Characterization of the mouse ClC-K1/Barttin chloride channel. Biochim. Biophys. Acta 1828, 2399–2409 (2013)

    Article  Google Scholar 

  26. Fahlke, C., Yu, H. T., Beck, C. L., Rhodes, T. H. & George, A. L. Jr. Pore-forming segments in voltage-gated chloride channels. Nature 390, 529–532 (1997)

    Article  ADS  CAS  Google Scholar 

  27. Accardi, A. & Pusch, M. Fast and slow gating relaxations in the muscle chloride channel CLC-1. J. Gen. Physiol. 116, 433–444 (2000)

    Article  CAS  Google Scholar 

  28. Estévez, R., Schroeder, B. C., Accardi, A., Jentsch, T. J. & Pusch, M. Conservation of chloride channel structure revealed by an inhibitor binding site in ClC-1. Neuron 38, 47–59 (2003)

    Article  Google Scholar 

  29. Waldegger, S. et al. Barttin increases surface expression and changes current properties of ClC-K channels. Pflugers Arch. 444, 411–418 (2002)

    Article  CAS  Google Scholar 

  30. Feng, L., Campbell, E. B. & MacKinnon, R. Molecular mechanism of proton transport in CLC Cl/H+ exchange transporters. Proc. Natl Acad. Sci. USA 109, 11699–11704 (2012)

    Article  ADS  CAS  Google Scholar 

  31. Markovic, S. & Dutzler, R. The structure of the cytoplasmic domain of the chloride channel ClC-Ka reveals a conserved interaction interface. Structure 15, 715–725 (2007)

    Article  CAS  Google Scholar 

  32. Robertson, J. L., Kolmakova-Partensky, L. & Miller, C. Design, function and structure of a monomeric ClC transporter. Nature 468, 844–847 (2010)

    Article  ADS  CAS  Google Scholar 

  33. Khantwal, C. M. et al. Revealing an outward-facing open conformational state in a CLC Cl/H+ exchange transporter. eLife 5, e11189 (2016)

    Article  Google Scholar 

  34. Miller, C. Open-state substructure of single chloride channels from Torpedo electroplax. Phil. Trans. R. Soc. Lond. B 299, 401–411 (1982)

    Article  ADS  CAS  Google Scholar 

  35. Duffield, M., Rychkov, G., Bretag, A. & Roberts, M. Involvement of helices at the dimer interface in ClC-1 common gating. J. Gen. Physiol. 121, 149–161 (2003)

    Article  CAS  Google Scholar 

  36. Gradogna, A., Fenollar-Ferrer, C., Forrest, L. R. & Pusch, M. Dissecting a regulatory calcium-binding site of CLC-K kidney chloride channels. J. Gen. Physiol. 140, 681–696 (2012)

    Article  CAS  Google Scholar 

  37. Jayaram, H., Robertson, J. L., Wu, F., Williams, C. & Miller, C. Structure of a slow CLC Cl/H+ antiporter from a cyanobacterium. Biochemistry 50, 788–794 (2011)

    Article  CAS  Google Scholar 

  38. Lim, H. H., Stockbridge, R. B. & Miller, C. Fluoride-dependent interruption of the transport cycle of a CLC Cl/H+ antiporter. Nature Chem. Biol. 9, 721–725 (2013)

    Article  CAS  Google Scholar 

  39. Bell, S. P., Curran, P. K., Choi, S. & Mindell, J. A. Site-directed fluorescence studies of a prokaryotic ClC antiporter. Biochemistry 45, 6773–6782 (2006)

    Article  CAS  Google Scholar 

  40. Basilio, D., Noack, K., Picollo, A. & Accardi, A. Conformational changes required for H+/Cl exchange mediated by a CLC transporter. Nature Struct. Mol. Biol . 21, 456–463 (2014)

    Article  CAS  Google Scholar 

  41. Jackson, M. R. et al. A preference for edgewise interactions between aromatic rings and carboxylate anions: the biological relevance of anion-quadrupole interactions. J. Phys. Chem. B 111, 8242–8249 (2007)

    Article  CAS  Google Scholar 

  42. Jardetzky, O. Simple allosteric model for membrane pumps. Nature 211, 969–970 (1966)

    Article  ADS  CAS  Google Scholar 

  43. Nguitragool, W. & Miller, C. Uncoupling of a CLC Cl/H+ exchange transporter by polyatomic anions. J. Mol. Biol. 362, 682–690 (2006)

    Article  CAS  Google Scholar 

  44. Accardi, A., Lobet, S., Williams, C., Miller, C. & Dutzler, R. Synergism between halide binding and proton transport in a CLC-type exchanger. J. Mol. Biol. 362, 691–699 (2006)

    Article  CAS  Google Scholar 

  45. Picollo, A., Malvezzi, M., Houtman, J. C. & Accardi, A. Basis of substrate binding and conservation of selectivity in the CLC family of channels and transporters. Nature Struct. Mol. Biol . 16, 1294–1301 (2009)

    Article  CAS  Google Scholar 

  46. Bergsdorf, E. Y., Zdebik, A. A. & Jentsch, T. J. Residues important for nitrate/proton coupling in plant and mammalian CLC transporters. J. Biol. Chem. 284, 11184–11193 (2009)

    Article  CAS  Google Scholar 

  47. Zifarelli, G. & Pusch, M. Conversion of the 2 Cl/1 H+ antiporter ClC-5 in a NO3/H+ antiporter by a single point mutation. EMBO J. 28, 175–182 (2009)

    Article  CAS  Google Scholar 

  48. De Angeli, A. et al. The nitrate/proton antiporter AtCLCa mediates nitrate accumulation in plant vacuoles. Nature 442, 939–942 (2006)

    Article  ADS  CAS  Google Scholar 

  49. Kim, J. H. et al. High cleavage efficiency of a 2A peptide derived from porcine teschovirus-1 in human cell lines, zebrafish and mice. PLoS One 6, e18556 (2011)

    Article  ADS  CAS  Google Scholar 

  50. Hofmann, C. et al. Efficient gene transfer into human hepatocytes by baculovirus vectors. Proc. Natl Acad. Sci. USA 92, 10099–10103 (1995)

    Article  ADS  CAS  Google Scholar 

  51. 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 

  52. Kirchhofer, A. et al. Modulation of protein properties in living cells using nanobodies. Nature Struct. Mol. Biol . 17, 133–138 (2010)

    Article  CAS  Google Scholar 

  53. Lim, H. H., Fang, Y. & Williams, C. High-efficiency screening of monoclonal antibodies for membrane protein crystallography. PLoS One 6, e24653 (2011)

    Article  ADS  CAS  Google Scholar 

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

    Article  Google Scholar 

  55. Grant, T. & Grigorieff, N. Measuring the optimal exposure for single particle cryo-EM using a 2.6 Å reconstruction of rotavirus VP6. eLife 4, e06980 (2015)

    Article  Google Scholar 

  56. Rohou, A. & Grigorieff, N. CTFFIND4: fast and accurate defocus estimation from electron micrographs. J. Struct. Biol. 192, 216–221 (2015)

    Article  Google Scholar 

  57. 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 

  58. Tang, G. et al. EMAN2: an extensible image processing suite for electron microscopy. J. Struct. Biol. 157, 38–46 (2007)

    Article  CAS  Google Scholar 

  59. Rubinstein, J. L. & Brubaker, M. A. Alignment of cryo-EM movies of individual particles by optimization of image translations. J. Struct. Biol. 192, 188–195 (2015)

    Article  Google Scholar 

  60. 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 

  61. Cardone, G., Heymann, J. B. & Steven, A. C. One number does not fit all: mapping local variations in resolution in cryo-EM reconstructions. J. Struct. Biol. 184, 226–236 (2013)

    Article  Google Scholar 

  62. Kucukelbir, A., Sigworth, F. J. & Tagare, H. D. Quantifying the local resolution of cryo-EM density maps. Nature Methods 11, 63–65 (2014)

    Article  CAS  Google Scholar 

  63. 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 

  64. Murshudov, G. N. et al. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr. D 67, 355–367 (2011)

    Article  CAS  Google Scholar 

  65. Brown, A. et al. Tools for macromolecular model building and refinement into electron cryo-microscopy reconstructions. Acta Crystallogr. D 71, 136–153 (2015)

    Article  CAS  Google Scholar 

  66. Nicholls, R. A., Long, F. & Murshudov, G. N. Low-resolution refinement tools in REFMAC5. Acta Crystallogr. D 68, 404–417 (2012)

    Article  CAS  Google Scholar 

  67. Fernández, I. S., Bai, X. C., Murshudov, G., Scheres, S. H. & Ramakrishnan, V. Initiation of translation by cricket paralysis virus IRES requires its translocation in the ribosome. Cell 157, 823–831 (2014)

    Article  Google Scholar 

  68. Ho, B. K. & Gruswitz, F. HOLLOW: generating accurate representations of channel and interior surfaces in molecular structures. BMC Struct. Biol. 8, 49 (2008)

    Article  Google Scholar 

  69. Chovancova, E. et al. CAVER 3.0: a tool for the analysis of transport pathways in dynamic protein structures. PLOS Comput. Biol. 8, e1002708 (2012)

    Article  CAS  Google Scholar 

  70. 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  ADS  CAS  Google Scholar 

  71. Lawrence, M. C. & Colman, P. M. Shape complementarity at protein/protein interfaces. J. Mol. Biol. 234, 946–950 (1993)

    Article  CAS  Google Scholar 

  72. Fialho, D. et al. Chloride channel myotonia: exon 8 hot-spot for dominant-negative interactions. Brain 130, 3265–3274 (2007)

    Article  CAS  Google Scholar 

  73. Vindas-Smith, R. et al. Identification and functional characterization of CLCN1 mutations found in nondystrophic myotonia patients. Hum. Mutat. 37, 74–83 (2016)

    Article  CAS  Google Scholar 

  74. Mailänder, V., Heine, R., Deymeer, F. & Lehmann-Horn, F. Novel muscle chloride channel mutations and their effects on heterozygous carriers. Am. J. Hum. Genet. 58, 317–324 (1996)

    PubMed  PubMed Central  Google Scholar 

  75. Trip, J. et al. In tandem analysis of CLCN1 and SCN4A greatly enhances mutation detection in families with non-dystrophic myotonia. Eur. J. Hum. Genet. 16, 921–929 (2008)

    Article  CAS  Google Scholar 

  76. Dupré, N. et al. Clinical, electrophysiologic, and genetic study of non-dystrophic myotonia in French-Canadians. Neuromuscul. Disord. 19, 330–334 (2009)

    Article  Google Scholar 

  77. Lorenz, C., Meyer-Kleine, C., Steinmeyer, K., Koch, M. C. & Jentsch, T. J. Genomic organization of the human muscle chloride channel CIC-1 and analysis of novel mutations leading to Becker-type myotonia. Hum. Mol. Genet. 3, 941–946 (1994)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Ebrahim at the Rockefeller University Cryo-EM Resource Center for help with microscope operation, staff at the Memorial Sloan Kettering Cancer Center Antibody & Bioresource Core Facility for hybridoma generation, Y. C. Hsiung for help with large-scale cell culture, members of the MacKinnon laboratory for helpful discussions, and J. Chen for critical reading of the manuscript. E.P. is supported by the Jane Coffin Childs Memorial Fund fellowship (#61-1513). R.M. is a Howard Hughes Medical Institute investigator.

Author information

Authors and Affiliations

Authors

Contributions

E.P. performed experiments. E.B.C. assisted in development of monoclonal antibodies. E.P. and R.M. designed experiments, analysed and interpreted results, and wrote the manuscript.

Corresponding author

Correspondence to Roderick MacKinnon.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks C. Miller, M. Pusch, S. Scheres and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Extended data figures and tables

Extended Data Figure 1 Functional characterization of the bovine CLC-K channel.

a, Representative two-electrode voltage clamp (TEVC) recordings of bovine CLC-K channel in X. laevis oocytes. Clamping voltages were from −60 to +60 mV (10-mV steps). be, IV curves of recordings in a. Shown are means and standard deviations (s.d.; error bars) of 5, 9, 10 and 11 independent oocyte recordings, respectively. f, IV curves of whole-cell patch recordings on Chinese hamster ovary (CHO) cells expressing CLC-K and barttin. The pipette and bath solutions contain 144 and 52 mM Cl, respectively. Shown are means and s.d. (error bars) of 3 or 5 independent recordings using different cells. g, Immunofluorescence staining of the bovine CLC-K channel expressed on the plasma membrane. CHO cells were transiently transfected with a green fluorescent protein (GFP)-tagged channel construct alone or together with barttin, and then cell-surface-targeted channels were probed by non-permeabilized immunofluorescence (IF) staining using monoclonal anti-CLC-K antibodies (clone 16E3), which specifically recognize an extracellular epitope of the CLC-K channel. Hoechst 33342 was used to stain nuclei. The same exposure parameters were used for the left and right panels. Shown are representative images of reproducible results. Scale bar, 10 μm.

Extended Data Figure 2 Cryo-EM image processing procedure.

a, A representative micrograph of the CLC-K–Fab complex (scale bar, 50 nm). Boxed regions (white squares) are magnified on the right to show representative particle images. Note that in the top/bottom views, Fab signals are evident while the channel part is barely visible because of low contrast (also see b). b, Images of selected two-dimensional classes from reference-free two-dimensional classification by RELION. Note that the distal half of the Fab fragment shows much flexibility, resulting in blurred features. Scale bar, 10 nm. c, Summary of the image processing procedure (see Methods).

Extended Data Figure 3 Three-dimensional reconstruction of CLC-K.

a, Angular distribution histogram of class 1 particle projections. b, Fourier shell correlation (FSC) of class 1 half maps before (black) and after (blue and red) masking. Two soft masks were used (see also c): one (red) including only the channel portion and the other (blue) including the channel and the variable domain (VH/VL) of the Fab fragments but excluding the constant domain (CH1/CL) of Fab. When a mask was used, the FSC curve was corrected for masking effects during the RELION postprocessing procedure (phase randomization above 7.8 Å). c, Masks used in FSC calculations (red and blue) in b are shown with the unsharpened, unfiltered map (grey) superimposed. A contour level of 0.7 was used for the surface representation of the masks. d, Local resolution map of the class 1 reconstruction estimated by the blocres program. Shown is the combined map, which is not sharpened or filtered. e, FSC between model and map of the class 1 particles. The black curve shows FSC between the final refined atomic model and the combined map that the model was refined against. The blue and red curves show FSC between the atomic model and the half map it was refined against (half 1) and FSC between the atomic model and the other half map it was not refined against (half 2), respectively (see also Methods). fj, As in ae, but with the class 2 structure. For the FSC calculations with the masks in g, the FSC curves were corrected for masking effects (phase randomization above 8.7 Å).

Extended Data Figure 4 EM density of the CLC-K channel.

EM density segments (mesh) of the class-1 CLC-K reconstruction are superimposed with the model in a ribbon representation. Numbers indicate amino acid positions of segments. The density map was sharpened with a B-factor of −100 Å2 and low-pass filtered at 3.7 Å. CTD, cytosolic domain. VL, variable domain of the IgG light chain. VH, variable domain of the IgG heavy chain. Note that a model for the distal half of the Fab fragment was not built because of poor density features in this region caused by its high flexibility (see also Extended Data Fig. 2b).

Extended Data Figure 5 Comparison between the class 1 and 2 structures of CLC-K.

a, The dimer interfaces (dashed boxes) of the class 1 and 2 structures were compared. Views from the extracellular side were aligned based on the upper (red) subunit. In addition, the right panel shows superposition of a class 1 monomer (grey) onto a class 2 monomer (red). b, N-terminal amphipathic helix of class 2 structure. A side view of the class 2 structure is shown in a ribbon representation superimposed with density (mesh) of the N-terminal helix. The two subunits are shown in pale green and blue. The TM helix αB, which is visible in both class 1 and 2 structures is shown in dark green. A 12-amino-acid amphipathic helix preceding αB, visible only in class 2, is shown in yellow. The density map was sharpened with a B-factor of −120 Å2 and low-pass filtered at 4.1 Å. The approximate membrane boundaries are indicated by grey lines.

Extended Data Figure 6 Structural features of the CLC-K channel.

a, Comparison between the CLC-K (class 1; grey and magenta) and EcCLC (yellow) structures. The TM domain of an EcCLC monomer was superposed onto that of CLC-K. α-Helices are represented as cylinders. b, The density of the extracellular loops αI–J of the CLC-K channel (class 1) is shown in blue and orange. The density of the remaining parts is shown in pale blue and yellow. In the top right panel, the model of one subunit is shown in the ribbon representation. αN and αF are highlighted, and Val166 and Tyr520 are shown in a ball-and-stick representation (green). The two ends of the αI–J link are indicated as cyan spheres with amino acid positions. The bottom panels show side views of one subunit with the αI–J link in blue density. The density map (5σ cut-off) was sharpened with a B-factor of −100 Å2 and low-pass filtered at 4.0 Å. c, As in the top left panel of b, but additionally showing a model of the αI–J link of EcCLC in a ball-and-stick representation (yellow). d, The same view as in the bottom right panel of b, but with superposition of an EcCLC model (pink; PDB accession 1OTS) onto the CLC-K model. A model for the αI–J linker of EcCLC is shown in yellow balls and sticks. e, Sequence alignment of the extracellular segments between TM helices αK and αM from various CLC proteins. The segments forming α-helices in the extracellular domain of bovine CLC-K are indicated by red coils. Red and blue dots indicate positions of mutations causing Bartter syndrome (CLC-Kb) and myotonia congenita (CLC-1), respectively. The A349D (ref. 5) mutation in CLC-Kb causes Bartter syndrome. The P408A (ref. 72), Q412P (ref. 73), F413C (ref. 4), A415V (ref. 74), E417G (ref. 75) or W433R (ref. 76) mutations in CLC-1 (corresponding to positions 342, 346, 347, 349, 351 and 367 of CLC-K, respectively) cause myotonia congenita. In addition, mutations at R438 (R438C in CLC-Kb or R496S in CLC-1), of which the side chain directly interacts with W367 (W433 in CLC-1) cause Bartter syndrome5 or myotonia congenita77 (see Fig. 4c).

Extended Data Figure 7 Structure of the ion transport pathway.

a, Stereo images of the atomic model of the CLC-K channel’s Cl-selective filter (class 1; light magenta) and comparisons with that of CmCLC (cyan) and EcCLC (yellow) are shown in a stick representation. In the cases of CmCLC and EcCLC, bound Cl ions are shown as spheres. Predicted polar interactions are indicated by grey dashed lines. b, EM density and model of the αC–D loop are shown for the class-2 CLC-K structure. Also, density and model were shown for the Tyr520 side chain. The density map (grey mesh) was sharpened with a B-factor of −120 Å2 and low-pass filtered at 4.1 Å.

Extended Data Figure 8 Density in the Scen Cl-binding site.

A view into Scen of CLC-K (from Sext towards the cytosol) is shown in a stereo diagram. Predicted polar interactions between bound Cl ions and the protein, including anion–quadrupole interactions, are indicated by green dashed lines. The shown EM density map is of the class 1 structure, sharpened with a B-factor of −100 Å2 and low-pass filtered at 3.7 Å. Density for protein (grey mesh) and possible ions (magenta mesh) are shown at contour levels of 5.8σ and 3.2σ, respectively.

Extended Data Figure 9 Surface electrostatic potential.

Surface electrostatics of the CLC-K channel (class 1) compared with the EcCLC and CmCLC transporters. Left panels show side views with one subunit in a surface representation and the other in a ribbon representation. Electrostatic potential is indicated by the colour scale (bottom left). Views from the extracellular side (blue arrow) and intracellular side (red arrow) are shown in the middle and right panels, respectively. The entrances to the Cl ion pathway are indicated by dashed circles. Mobile monovalent cation and anion species at 0.15 M each were included in the electrostatic potential calculations.

Extended Data Table 1 Model refinement statistics

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Park, E., Campbell, E. & MacKinnon, R. Structure of a CLC chloride ion channel by cryo-electron microscopy. Nature 541, 500–505 (2017). https://doi.org/10.1038/nature20812

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature20812

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