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:

Stochastic transport through carbon nanotubes in lipid bilayers and live cell membranes

Abstract

There is much interest in developing synthetic analogues of biological membrane channels1 with high efficiency and exquisite selectivity for transporting ions and molecules. Bottom-up2 and top-down3 methods can produce nanopores of a size comparable to that of endogenous protein channels, but replicating their affinity and transport properties remains challenging. In principle, carbon nanotubes (CNTs) should be an ideal membrane channel platform: they exhibit excellent transport properties4,5,6,7,8 and their narrow hydrophobic inner pores mimic structural motifs typical of biological channels1. Moreover, simulations predict that CNTs with a length comparable to the thickness of a lipid bilayer membrane can self-insert into the membrane9,10. Functionalized CNTs have indeed been found to penetrate lipid membranes and cell walls11,12, and short tubes have been forced into membranes to create sensors13, yet membrane transport applications of short CNTs remain underexplored. Here we show that short CNTs spontaneously insert into lipid bilayers and live cell membranes to form channels that exhibit a unitary conductance of 70–100 picosiemens under physiological conditions. Despite their structural simplicity, these ‘CNT porins’ transport water, protons, small ions and DNA, stochastically switch between metastable conductance substates, and display characteristic macromolecule-induced ionic current blockades. We also show that local channel and membrane charges can control the conductance and ion selectivity of the CNT porins, thereby establishing these nanopores as a promising biomimetic platform for developing cell interfaces, studying transport in biological channels, and creating stochastic sensors.

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: Synthesis, characterization, and bulk transport properties of CNT porins.
Figure 2: Transport through individual CNT porins.
Figure 3: DNA translocation through CNT ion channels.
Figure 4: CNT porin incorporation into live cell membranes.

Similar content being viewed by others

References

  1. Sui, H., Han, B.-G., Lee, J. K., Walian, P. & Jap, B. K. Structural basis of water-specific transport through the AQP1 water channel. Nature 414, 872–878 (2001)

    Article  ADS  CAS  Google Scholar 

  2. Langecker, M. et al. Synthetic lipid membrane channels formed by designed DNA nanostructures. Science 338, 932–936 (2012)

    Article  ADS  CAS  Google Scholar 

  3. Li, J. et al. Ion-beam sculpting at nanometre length scales. Nature 412, 166–169 (2001)

    Article  ADS  CAS  Google Scholar 

  4. Hinds, B. J. et al. Aligned multiwalled carbon nanotube membranes. Science 303, 62–65 (2004)

    Article  ADS  CAS  Google Scholar 

  5. Liu, H. et al. Translocation of single-stranded DNA through single-walled carbon nanotubes. Science 327, 64–67 (2010)

    Article  ADS  CAS  Google Scholar 

  6. Lee, C. Y., Choi, W., Han, J.-H. & Strano, M. S. Coherence resonance in a single-walled carbon nanotube ion channel. Science 329, 1320–1324 (2010)

    Article  ADS  CAS  Google Scholar 

  7. Zimmerli, U. & Koumoutsakos, P. Simulations of electrophoretic RNA transport through transmembrane carbon nanotubes. Biophys. J. 94, 2546–2557 (2008)

    Article  CAS  Google Scholar 

  8. Hummer, G., Rasaiah, J. C. & Noworyta, J. P. Water conduction through the hydrophobic channel of a carbon nanotube. Nature 414, 188–190 (2001)

    Article  ADS  CAS  Google Scholar 

  9. Lopez, C. F., Nielsen, S. O., Moore, P. B. & Klein, M. L. Understanding nature's design for a nanosyringe. Proc. Natl Acad. Sci. USA 101, 4431–4434 (2004)

    Article  ADS  CAS  Google Scholar 

  10. Dutt, M., Nayhouse, M. J., Kuksenok, O., Little, S. R. & Balazs, A. C. Interactions of end-functionalized nanotubes with lipid vesicles: spontaneous insertion and nanotube self-organization. Curr. Nanosci. 7, 699–715 (2011)

    Article  ADS  CAS  Google Scholar 

  11. Lacerda, L. et al. How do functionalized carbon nanotubes land on, bind to and pierce through model and plasma membranes. Nanoscale 5, 10242–10250 (2013)

    Article  ADS  CAS  Google Scholar 

  12. Kam, N. W. S., O’Connell, M., Wisdom, J. A. & Dai, H. Carbon nanotubes as multifunctional biological transporters and near-infrared agents for selective cancer cell destruction. Proc. Natl Acad. Sci. USA 102, 11600–11605 (2005)

    Article  ADS  CAS  Google Scholar 

  13. Liu, L., Yang, C., Zhao, K., Li, J. & Wu, H.-C. Ultrashort single-walled carbon nanotubes in a lipid bilayer as a new nanopore sensor. Nature Commun. 4, http://dx.doi.org/10.1038/ncomms3989 (2013)

  14. Sun, X. et al. Optical properties of ultrashort semiconducting single-walled carbon nanotube capsules down to sub-10 nm. J. Am. Chem. Soc. 130, 6551–6555 (2008)

    Article  CAS  Google Scholar 

  15. Le Duc, Y. et al. Imidazole-quartet water and proton dipolar channels. Angew. Chem. Int. Ed. 50, 11366–11372 (2011)

    Article  CAS  Google Scholar 

  16. Fornasiero, F. et al. Ion exclusion by sub 2-nm carbon nanotube pores. Proc. Natl Acad. Sci. USA 105, 17250–17255 (2008)

    Article  ADS  CAS  Google Scholar 

  17. Walther, J. H., Ritos, K., Cruz-Chu, E. R., Megaridis, C. M. & Koumoutsakos, P. Barriers to superfast water transport in carbon nanotube membranes. Nano Lett. 13, 1910–1914 (2013)

    Article  ADS  CAS  Google Scholar 

  18. Gu, L.-Q. & Bayley, H. Interaction of the noncovalent molecular adapter, β-cyclodextrin, with the staphylococcal α-hemolysin pore. Biophys. J. 79, 1967–1975 (2000)

    Article  CAS  Google Scholar 

  19. Nestorovich, E. M., Rostovtseva, T. K. & Bezrukov, S. M. Residue ionization and ion transport through OmpF channels. Biophys. J. 85, 3718–3729 (2003)

    Article  CAS  Google Scholar 

  20. Powell, M. R. et al. Nanoprecipitation-assisted ion current oscillations. Nature Nanotechnol. 3, 51–57 (2007)

    Article  ADS  Google Scholar 

  21. Powell, M. R., Cleary, L., Davenport, M., Shea, K. J. & Siwy, Z. S. Electric-field-induced wetting and dewetting in single hydrophobic nanopores. Nature Nanotechnol. 6, 798–802 (2011)

    Article  ADS  CAS  Google Scholar 

  22. Lev, A. A. et al. Rapid switching of ion current in narrow pores: implications for biological ion channels. Proc. R. Soc. Lond. B 252, 187–192 (1993)

    Article  ADS  CAS  Google Scholar 

  23. Shimizu, S. et al. Stochastic pore blocking and gating in PDMS–glass nanopores from vapor–liquid phase transitions. J. Phys. Chem. C 117, 9641–9651 (2013)

    Article  CAS  Google Scholar 

  24. Buyukdagli, S., Manghi, M. & Palmeri, J. Ionic capillary evaporation in weakly charged nanopores. Phys. Rev. Lett. 105, 158103 (2010)

    Article  ADS  Google Scholar 

  25. Kasianowicz, J. J., Brandin, E., Branton, D. & Deamer, D. W. Characterization of individual polynucleotide molecules using a membrane channel. Proc. Natl Acad. Sci. USA 93, 13770–13773 (1996)

    Article  ADS  CAS  Google Scholar 

  26. Hall, A. R. et al. Hybrid pore formation by directed insertion of α-haemolysin into solid-state nanopores. Nature Nanotechnol. 5, 874–877 (2010)

    Article  ADS  CAS  Google Scholar 

  27. Venkatesan, B. M. & Bashir, R. Nanopore sensors for nucleic acid analysis. Nature Nanotechnol. 6, 615–624 (2011)

    Article  ADS  CAS  Google Scholar 

  28. Majumder, M., Chopra, N. & Hinds, B. J. Effect of tip functionalization on transport through vertically oriented carbon nanotube membranes. J. Am. Chem. Soc. 127, 9062–9070 (2005)

    Article  CAS  Google Scholar 

  29. Haque, F., Geng, J., Montemagno, C. & Guo, P. Incorporation of a viral DNA-packaging motor channel in lipid bilayers for real-time, single-molecule sensing of chemicals and double-stranded DNA. Nature Protocols 8, 373–392 (2013)

    Article  CAS  Google Scholar 

  30. Neher, E., Sandblom, J. & Eisenman, G. Ionic selectivity, saturation, and block in gramicidin A channels. J. Membr. Biol. 40, 97–116 (1978)

    Article  CAS  Google Scholar 

  31. Frolov, V. A., Lizunov, V. A., Dunina-Barkovskaya, A. Y., Samsonov, A. V. & Zimmerberg, J. Shape bistability of a membrane neck: a toggle switch to control vesicle content release. Proc. Natl Acad. Sci. USA 100, 8698–8703 (2003)

    Article  ADS  CAS  Google Scholar 

  32. Shnyrova, A. V. et al. Geometric catalysis of membrane fission driven by flexible dynamin rings. Science 339, 1433–1436 (2013)

    Article  ADS  CAS  Google Scholar 

  33. Edelstein, A., Amodaj, N., Hoover, K., Vale, R. & Stuurman, N. Computer control of microscopes using µManager. Curr. Protoc. Molec. Biol. 92, 14.20.1–14.20.17 (2010)

    Article  Google Scholar 

Download references

Acknowledgements

Parts of this work were supported by the US Department of Energy, Office of Basic Energy Sciences, Division of Materials Sciences and Engineering (characterization and transport studies), and the LDRD programme at LLNL, 12-ERD-073 (synthesis). R.T. acknowledges support from the LSP programme at LLNL. D.M. acknowledges support from an ROTC summer fellowship. V.A.F. acknowledges partial support by the Spanish Ministry of Economy and Competitiveness, grant BFU2012-34885, co-financed with European FEDER funds, and the Basque Government, grant IE12-332. Work at LLNL was performed under the auspices of the US Department of Energy under contract DE-AC52-07NA27344. Work at the Molecular Foundry was supported by the Office of Science, Office of Basic Energy Sciences, of the US Department of Energy under contract DE-AC02-05CH11231.

Author information

Authors and Affiliations

Authors

Contributions

J.G. performed and analysed (with D.M.) the planar lipid bilayer transport measurements; K.K. performed bulk transport studies; J.Z., K.K., J.G. and R.T. performed the synthesis and purification, K.R.C. performed AFM analysis, Y.M.W., L.R.C., F.I.A. and K.K. performed TEM analysis; L.R.C. and F.I.A. performed cryo-TEM analysis; A.N. conceived and directed the research, and wrote the manuscript draft; A.E., A.V.S. and V.A.F. designed the cell and GUV reconstitution experiments, and A.E. and A.V.S. performed them. All authors contributed to the data analysis, discussion, and manuscript preparation.

Corresponding author

Correspondence to Aleksandr Noy.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 TEM images of isolated CNTs.

ac, Purified CNTs before cutting (a and b, conventional TEM, c imaged on a cryo stage; all air-dried samples). d, Magnified image of one CNT from c. eg, CNTs mixed with lipid and lightly sonicated to aid nanotube dispersion. hm, Isolated CNT porins after the cutting procedure.

Extended Data Figure 2 Length distributions of CNT porins measured from TEM images.

a, CNT porin sample after cutting and purification procedures (n = 65). b, Cut and purified sample after passing through a 100 nm pore filter membrane (n = 36). c, As b but after passing through a 50 nm pore filter membrane (n = 156). µ indicates the mean CNT porin length.

Extended Data Figure 3 Characterization of CNT porins.

a, Histogram of the diameters of short CNT fragments measured with AFM. Most CNT porins show diameters between 1 and 2 nm. Inset, high-magnification AFM image of a single CNT on a bare mica surface. b, Histogram of the diameters of CNT porins inserted into the lipid vesicles measured by cryo-TEM. c, Raman spectrum of the short-CNT/lipid complex after 16 h of sonication-assisted cutting. Inset, magnified view of the radial breathing mode region of the CNT spectrum (100–300 cm−1).

Extended Data Figure 4 Cryo-TEM images of CNT–liposome complexes.

Original large field-of-view images (main panels); insets, magnified views of the selected structures, with false-colour lines added to highlight the lipid bilayer (red) and CNT porins (blue).

Extended Data Figure 5 High magnification cryo-TEM imaged of CNT–liposome complexes.

Panels (left to right) show unprocessed, image-processed, and colourized unprocessed examples. See Methods for details of image processing.

Extended Data Figure 6 Single CNT porin conductance at different electrolyte concentrations.

Plots of the average single CNT porin conductance at different values of background electrolyte (KCl) concentrations at pH 7 (red circles) and pH 2 (blue squares). The slopes of the linear fit through the data (dashed lines) correspond to 0.66 nS M−1 at pH 7 and 0.36 nS M−1 at pH 2. Error bars, s.d.; n values as follows (pH = 7: 0.15 M, n = 6; 0.5 M, n = 161; 1 M, n = 39; 2 M, n = 8. pH = 2: 0.5 M, n = 18; 1 M, n = 3; 2 M, n = 36).

Extended Data Figure 7 Single CNT porin conductance at pH 2.

a, Representative conductance traces showing individual CNT porin incorporation at pH 2 and 2 M KCl background electrolyte concentration. Traces i–iv represent incorporation from 4 independent experiments. b, Histogram of conductance values measured for 109 individual CNT channel incorporation events (n = 109). Solid blue line corresponds to the fit of the data to a distribution expected for multiple channel incorporation, where the position and width of the second peak are determined from the position, μ (0.36 ± 0.006 nS), and width, σ (0.10 ± 0.008 nS), of the first peak as 2μ and . c, Representative conductance traces showing individual CNT porin incorporation at pH 2 and 0.5 M KCl background electrolyte. Traces i–iv represent incorporation from 4 independent experiments. Control trace shows a conductance trace collected at the same conditions without adding CNT porins.

Extended Data Figure 8 Representative traces and corresponding current level histograms of CNT porin incorporation, gating and DNA translocation in lipid bilayers at pH 7.

a, Control conductance trace recorded in the absence of CNT porin. Although only the first 5 min are shown, the current level remained stable for up to 80 min (n = 3 × 106). b, c, Conductance traces and histograms showing single CNT porin incorporation events and subsequent current fluctuations between gating sub-levels (n > 5 × 104). d, Trace and conductance value histogram showing CNT porin incorporation event and subsequent two DNA translocation events (indicated by black arrows). Here n = 3.6 × 105. The peaks on the histogram correspond to 0.01 nS for pre-incorporation baseline, 0.22 nS for DNA blockade, and 0.67 nS for open pore conductance, which gives the DNA blockade of 0.45 nS and a blockade ratio of 67%. Dashed lines are added to all traces as a guide to the eye.

Extended Data Figure 9 CNT porin conductance in cellular membranes.

a, Schematic of the on-cell patch clamp measurement. b, c, On-cell conductance measurements demonstrate no channel activity (b) and appearance of a small flickering channel (c) in control experiments with HEK293T cells. Holding potentials were −60 mV (b) and +60 mV (c), the time order is from left to right, top to bottom. d, In the presence of CNT porin (0.15% dilution of the stock), large channel activity is detected, holding potential −60 mV. e, Metastable conductance sub-states of 50 pS detected with CNT porin incorporated into the plasma membrane of CHO cell, holding potential −60 mV. f, Mean patch conductance (on cell configuration, 0.15% CNT stock solution in the patch pipette) detected in CHO and HEK293T cells at positive and negative holding potentials. Error bar, s.d.; 4 cells). In 70% of HEK293T cells and 95% of CHO cells we did not observe significant dependence of the conductance on the holding potential; only those cells were used for the conductance analysis.

Extended Data Figure 10 CNT porin conductance in patches isolated from planar lipid bilayers and giant unilamellar vesicles.

a, Histogram of conductance values measured for CNT porin incorporation into a planar lipid bilayer mimicking the composition of a charged plasma membrane (red bars, n = 681, 5 membranes, 3 controls without CNT porins showed no events) and a similar lipid bilayer without charged species (black bars, n = 540, 5 membranes, 3 controls without CNT porins showed no events). The background electrolyte concentration was kept at 150 mM. Solid lines correspond to the fit of the data to a distribution expected for multiple channel incorporation. The primary peak positions are 65.1 ± 0.4 pS (charged bilayer) and 92.5 ± 0.6 pS (uncharged bilayer). b, Histogram of conductance values measured for CNT porins reconstituted into giant unilamellar vesicles (GUVs) mimicking the composition of charged plasma membrane (red bars, n = 40, 3 membranes), Solid lines correspond to the fit of the data to a distribution expected for multiple channel incorporation with the primary conductance peak at 60 ± 2 pS. c, Representative control experiment demonstrating lack of spontaneous pore formation in the bilayer patch. d, e, Patch-clamp measurements of the CNT porin incorporation events in planar lipid bilayer (d) and GUV membrane (e).

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Geng, J., Kim, K., Zhang, J. et al. Stochastic transport through carbon nanotubes in lipid bilayers and live cell membranes. Nature 514, 612–615 (2014). https://doi.org/10.1038/nature13817

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13817

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