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:

Electrical bioadhesive interface for bioelectronics

Abstract

Reliable functions of bioelectronic devices require conformal, stable and conductive interfaces with biological tissues. Integrating bioelectronic devices with tissues usually relies on physical attachment or surgical suturing; however, these methods face challenges such as non-conformal contact, unstable fixation, tissue damage, and/or scar formation. Here, we report an electrical bioadhesive (e-bioadhesive) interface, based on a thin layer of a graphene nanocomposite, that can provide rapid (adhesion formation within 5 s), robust (interfacial toughness >400 J m−2) and on-demand detachable integration of bioelectronic devices on diverse wet dynamic tissues. The electrical conductivity (>2.6 S m−1) of the e-bioadhesive interface further allows bidirectional bioelectronic communications. We demonstrate biocompatibility, applicability, mechanical and electrical stability, and recording and stimulation functionalities of the e-bioadhesive interface based on ex vivo porcine and in vivo rat models. These findings offer a promising strategy to improve tissue–device integration and enhance the performance of biointegrated electronic devices.

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: Design and mechanism of the e-bioadhesive interface.
Fig. 2: Mechanical properties of the e-bioadhesive interface.
Fig. 3: Electrical properties of the e-bioadhesive interface.
Fig. 4: In vitro and in vivo biocompatibility of the e-bioadhesive interface.
Fig. 5: In vivo recording and stimulation via the e-bioadhesive interface.

Similar content being viewed by others

Data availability

All relevant data that support the findings of this study are available in the article and its supplementary files. Source data of plots are provided with this paper. Source data are provided with this paper.

Code availability

No custom code is used in this study.

References

  1. Rogers, J. A., Someya, T. & Huang, Y. Materials and mechanics for stretchable electronics. Science 327, 1603–1607 (2010).

    CAS  Google Scholar 

  2. Someya, T., Bao, Z. & Malliaras, G. G. The rise of plastic bioelectronics. Nature 540, 379–385 (2016).

    CAS  Google Scholar 

  3. Feiner, R. & Dvir, T. Tissue–electronics interfaces: from implantable devices to engineered tissues. Nat. Rev. Mater. 3, 17076 (2018).

    CAS  Google Scholar 

  4. Schiavone, G. & Lacour, S. P. Conformable bioelectronic interfaces: mapping the road ahead. Sci. Transl. Med. 11, eaaw5858 (2019).

    Google Scholar 

  5. Yuk, H., Lu, B. & Zhao, X. Hydrogel bioelectronics. Chem. Soc. Rev. 48, 1642–1667 (2019).

    CAS  Google Scholar 

  6. Zhang, Y. et al. Climbing-inspired twining electrodes using shape memory for peripheral nerve stimulation and recording. Sci. Adv. 5, eaaw1066 (2019).

    CAS  Google Scholar 

  7. Kim, D.-H. et al. Dissolvable films of silk fibroin for ultrathin conformal bio-integrated electronics. Nat. Mater. 9, 511–517 (2010).

    CAS  Google Scholar 

  8. Park, J. et al. Electromechanical cardioplasty using a wrapped elasto-conductive epicardial mesh. Sci. Transl. Med. 8, 344ra386 (2016).

    Google Scholar 

  9. Jeong, J. W. et al. Materials and optimized designs for human–machine interfaces via epidermal electronics. Adv. Mater. 25, 6839–6846 (2013).

    CAS  Google Scholar 

  10. Sim, K. et al. Metal oxide semiconductor nanomembrane-based soft unnoticeable multifunctional electronics for wearable human–machine interfaces. Sci. Adv. 5, eaav9653 (2019).

    CAS  Google Scholar 

  11. Kim, D.-H. et al. Epidermal electronics. Science 333, 838–843 (2011).

    CAS  Google Scholar 

  12. Kaltenbrunner, M. et al. An ultra-lightweight design for imperceptible plastic electronics. Nature 499, 458 (2013).

    CAS  Google Scholar 

  13. Lee, S. et al. A strain-absorbing design for tissue–machine interfaces using a tunable adhesive gel. Nat. Commun. 5, 5898 (2014).

    CAS  Google Scholar 

  14. Ouyang, H. et al. Symbiotic cardiac pacemaker. Nat. Commun. 10, 1821 (2019).

    Google Scholar 

  15. Boutry, C. M. et al. Biodegradable and flexible arterial-pulse sensor for the wireless monitoring of blood flow. Nat. Biomed. Eng. 3, 47–57 (2019).

    CAS  Google Scholar 

  16. Koo, J. et al. Wireless bioresorbable electronic system enables sustained nonpharmacological neuroregenerative therapy. Nat. Med. 24, 1830–1836 (2018).

    CAS  Google Scholar 

  17. Boutry, C. M. et al. A stretchable and biodegradable strain and pressure sensor for orthopaedic application. Nat. Electron. 1, 314–321 (2018).

    Google Scholar 

  18. Chang, E. I. et al. Vascular anastomosis using controlled phase transitions in poloxamer gels. Nat. Med. 17, 1147–1152 (2011).

    CAS  Google Scholar 

  19. Minev, I. R. et al. Electronic dura mater for long-term multimodal neural interfaces. Science 347, 159–163 (2015).

    CAS  Google Scholar 

  20. Formento, E. et al. Electrical spinal cord stimulation must preserve proprioception to enable locomotion in humans with spinal cord injury. Nat. Neurosci. 21, 1728–1741 (2018).

    CAS  Google Scholar 

  21. Roche, E. T. et al. Soft robotic sleeve supports heart function. Sci. Transl. Med. 9, eaaf3925 (2017).

    Google Scholar 

  22. Whyte, W. et al. Sustained release of targeted cardiac therapy with a replenishable implanted epicardial reservoir. Nat. Biomed. Eng. 2, 416–428 (2018).

    CAS  Google Scholar 

  23. Nido, P. D. & Goldman, B. S. Temporary epicardial pacing after open heart surgery: complications and prevention. J. Card. Surg. 4, 99–103 (1989).

    Google Scholar 

  24. Yang, S. et al. “Cut-and-paste” manufacture of multiparametric epidermal sensor systems. Adv. Mater. 27, 6423–6430 (2015).

    CAS  Google Scholar 

  25. Koh, A. et al. A soft, wearable microfluidic device for the capture, storage, and colorimetric sensing of sweat. Sci. Transl. Med. 8, 366ra165 (2016).

    Google Scholar 

  26. Liao, M. et al. Wearable, healable, and adhesive epidermal sensors assembled from mussel‐inspired conductive hybrid hydrogel framework. Adv. Funct. Mater. 27, 1703852 (2017).

    Google Scholar 

  27. Liang, S. et al. Paintable and rapidly bondable conductive hydrogels as therapeutic cardiac patches. Adv. Mater. 30, 1704235 (2018).

    Google Scholar 

  28. Sekitani, T. et al. Ultraflexible organic amplifier with biocompatible gel electrodes. Nat. Commun. 7, 11425 (2016).

    CAS  Google Scholar 

  29. Yamagishi, K. et al. Tissue-adhesive wirelessly powered optoelectronic device for metronomic photodynamic cancer therapy. Nat. Biomed. Eng. 3, 27–36 (2019).

    CAS  Google Scholar 

  30. Keplinger, C. et al. Stretchable, transparent, ionic conductors. Science 341, 984–987 (2013).

    CAS  Google Scholar 

  31. Li, J. et al. Tough adhesives for diverse wet surfaces. Science 357, 378–381 (2017).

    CAS  Google Scholar 

  32. Rose, S. et al. Nanoparticle solutions as adhesives for gels and biological tissues. Nature 505, 382–385 (2014).

    CAS  Google Scholar 

  33. Liu, J. et al. Intrinsically stretchable electrode array enabled in vivo electrophysiological mapping of atrial fibrillation at cellular resolution. Proc. Natl Acad. Sci. USA 117, 14769–14778 (2020).

    CAS  Google Scholar 

  34. Yuk, H. et al. Dry double-sided tape for adhesion of wet tissues and devices. Nature 575, 169–174 (2019).

    CAS  Google Scholar 

  35. Yang, J.-H. & Lee, Y.-D. Highly electrically conductive rGO/PVA composites with a network dispersive nanostructure. J. Mater. Chem. 22, 8512–8517 (2012).

    CAS  Google Scholar 

  36. Liu, J. et al. Fatigue-resistant adhesion of hydrogels. Nat. Commun. 11, 1–9 (2020).

    CAS  Google Scholar 

  37. Mao, X., Yuk, H. & Zhao, X. Hydration and swelling of dry polymers for wet adhesion. J. Mech. Phys. Solids 137, 103863 (2020).

    CAS  Google Scholar 

  38. Liu, J. et al. Triggerable tough hydrogels for gastric resident dosage forms. Nat. Commun. 8, 124 (2017).

    Google Scholar 

  39. Li, W. et al. Tough bonding, on-demand debonding, and facile rebonding between hydrogels and diverse metal surfaces. Adv. Mater. 31, 1904732 (2019).

    CAS  Google Scholar 

  40. Chen, X., Yuk, H., Wu, J., Nabzdyk, C. S. & Zhao, X. Instant tough bioadhesive with triggerable benign detachment. Proc. Natl Acad. Sci. USA 117, 15497–15503 (2020).

    CAS  Google Scholar 

  41. Kashyap, S., Pratihar, S. K. & Behera, S. K. Strong and ductile graphene oxide reinforced PVA nanocomposites. J. Alloy. Compd. 684, 254–260 (2016).

    CAS  Google Scholar 

  42. Gabriel, C., Peyman, A. & Grant, E. H. Electrical conductivity of tissue at frequencies below 1 MHz. Phys. Med. Biol. 54, 4863 (2009).

    CAS  Google Scholar 

  43. Cogan, S. F. Neural stimulation and recording electrodes. Annu. Rev. Biomed. Eng. 10, 275–309 (2008).

    CAS  Google Scholar 

  44. Liu, Y. et al. Soft and elastic hydrogel-based microelectronics for localized low-voltage neuromodulation. Nat. Biomed. Eng. 3, 58–68 (2019).

    CAS  Google Scholar 

  45. Zheng, H. et al. A shape-memory and spiral light-emitting device for precise multisite stimulation of nerve bundles. Nat. Commun. 10, 2790 (2019).

    Google Scholar 

  46. Gorgieva, S. & Kokol, V. Preparation, characterization, and in vitro enzymatic degradation of chitosan‐gelatine hydrogel scaffolds as potential biomaterials. J. Biomed. Mater. Res. A 100, 1655–1667 (2012).

    Google Scholar 

  47. Park, S. et al. Self-powered ultra-flexible electronics via nano-grating-patterned organic photovoltaics. Nature 561, 516–521 (2018).

    CAS  Google Scholar 

  48. Lee, S. et al. Ultrasoft electronics to monitor dynamically pulsing cardiomyocytes. Nat. Nanotechnol. 14, 156–160 (2019).

    CAS  Google Scholar 

  49. Webb, R. C. et al. Ultrathin conformal devices for precise and continuous thermal characterization of human skin. Nat. Mater. 12, 938–944 (2013).

    CAS  Google Scholar 

  50. Xu, L. et al. 3D multifunctional integumentary membranes for spatiotemporal cardiac measurements and stimulation across the entire epicardium. Nat. Commun. 5, 3329 (2014).

    Google Scholar 

  51. Kim, D.-H. et al. Materials for multifunctional balloon catheters with capabilities in cardiac electrophysiological mapping and ablation therapy. Nat. Mater. 10, 316–323 (2011).

    CAS  Google Scholar 

  52. Gutruf, P. et al. Wireless, battery-free, fully implantable multimodal and multisite pacemakers for applications in small animal models. Nat. Commun. 10, 5742 (2019).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank C.S. Nabzdyk at Mayo Clinic for the insightful discussions and suggestions, R. Bronson at Harvard Medical School for the histological analyses and D. Yang at MIT for help with the charge injection capacity tests. This work is supported by Centers for Mechanical Engineering Research and Education at MIT and SUSTech (MechERE Centers at MIT and SUSTech), the Innovation Committee of Shenzhen Municipality (JCYJ20170817111714314, C.F.G.), the National Natural Science Foundation of China (numbers U1613204 and 51771089, C.F.G.), the Science Technology the Shenzhen Sci-Tech Fund (KYTDPT20181011104007, C.F.G.) and the Guangdong Innovative and Entrepreneurial Research Team Program (2016ZT06G587, C.F.G.). H.Y. acknowledges financial support from a Samsung Scholarship.

Author information

Authors and Affiliations

Authors

Contributions

H.Y., J.D. and X.Z. conceived the idea. J.D., H.Y., C.F.G. and X.Z. designed the study. J.D. and H.Y. developed the materials and methods for the e-bioadhesive interface. J.D., H.Y., J.W. and X.C. designed and performed the in vitro and ex vivo experiments. H.Y., J.W., C.E.V., J.D. and E.T.R. designed the in vivo experiments. J.W., H.Y., C.E.V. and J.D. performed the in vivo experiments. J.D., H.Y., C.F.G. and X.Z. analysed the data and wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Chuan Fei Guo or Xuanhe Zhao.

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

Extended Data Fig. 1 Rapid and robust adhesion of a device on wet tissue surface by the e-bioadhesive interface.

a, Overall process of applying a device with the e-bioadhesive interface to an ex vivo porcine heart. b, Robust integration of the device to the wet heart surface.

Extended Data Fig. 2 Amine coupling of the e-bioadhesive interface.

a, Schematic illustration of covalent coupling of fluorescein with the e-bioadhesive interface by reaction between NHS ester groups in the e-bioadhesive interface and the primary amine groups in 6-amino fluorescein. b, c, Fluorescence microscopic images of the e-bioadhesive interface with (b) and without (c) NHS ester. 3 independent experiments were conducted with similar results.

Extended Data Fig. 3 Surface amine functionalization of various device materials.

a, Schematic illustration for primary amine functionalization of silicon and PDMS. b, Schematic illustration for primary amine functionalization of gold. c, Schematic illustration for primary amine functionalization of polyimide. d, Schematic illustration for primary amine functionalization of polycarbonate.

Extended Data Fig. 4 Schematic illustrations for the overall application process and mechanism of the e-bioadhesive interface.

The e-bioadhesive interface is assembled with bioelectronic devices to endow the ability to adhere to and electrically communicate with wet tissue surfaces. Upon contact with the wet tissue surface, the e-bioadhesive interface quickly absorbs the interfacial water and dry the tissue surface. The carboxylic acid groups in the e-bioadhesive interface form temporary physical crosslinks by hydrogen bonds and/or electrostatic interactions. The NHS ester groups in the e-bioadhesive interface subsequently form covalent crosslinks with the primary amine groups on the tissue surface and provide stable long-term tissue-device integration.

Extended Data Fig. 5 On-demand detachment of the e-bioadhesive interface.

a, Schematic illustrations for on-demand detachment of the e-bioadhesive interface by the triggering solution. b, Schematic illustrations for chemistry of the on-demand cleaving of hydrogen bonds between the e-bioadhesive interface and the tissue surface. c, Schematic illustrations for the on-demand cleaving of covalent disulfide bonds between the e-bioadhesive interface and the tissue surface.

Extended Data Fig. 6 Evaluation of compressive narrowing of carotid artery and sciatic nerve by the e-bioadhesive interface.

a, Schematic illustrations of a measurement setup for the potential compressive narrowing of tissues by the e-bioadhesive interface. b, Outer diameter of bare carotid artery, carotid artery with the dry and fully swollen e-bioadhesive interfaces. c, Outer diameter of bare sciatic nerve, sciatic nerve with the dry and fully swollen e-bioadhesive interfaces. Values in b,c represent the mean and the standard deviation (n = 4 independent samples). Statistical significance and P values are determined by two-sided Student t-test; ns, not significant.

Source data

Extended Data Fig. 7 Adhesion performance of device integration to wet tissues by physical attachment.

a, Interfacial toughness between various tissues and polyimide substrates adhered by the e-bioadhesive interface and physical attachment. b, Shear strength between various tissues and polyimide substrates adhered by the e-bioadhesive interface and physical attachment. Values in a, b represent the mean and the standard deviation (n = 3 independent samples). Statistical significance and P values are determined by two-sided Student t-test; *** p ≤ 0.001; **** p ≤ 0.0001.

Source data

Extended Data Fig. 8 Immunohistochemistry analysis of CD3, CD68, Collagen-I, and α-SMA expression.

Representative immunohistochemistry images of CD3, CD68, Collagen-I, and α-SMA for gold electrode with and without the e-bioadhesive interface after rat subcutaneous implantation for 14 days, respectively. 5 independent experiments were conducted with similar results.

Extended Data Fig. 9 Stability of in vivo performance.

Conductivity, impedance, and interfacial toughness of two gold electrodes adhered by the e-bioadhesive interface after 1 day and 14 days of incubation in PBS, and 14 days of in vivo implantation in rat dorsal subcutaneous pockets. Values represent the mean and the standard deviation (n = 3 independent samples). Statistical significance and P values are determined by two-sided Student t-test; ns, not significant.

Source data

Extended Data Table 1 Comparison of various types of bioelectronic devices for wet tissues and organs

Supplementary information

Supplementary Information

Supplementary Figs. 1–11, discussion and Table 1.

Reporting Summary

Supplementary Video 1

Application of LED circuits with and without the e-bioadhesive interface on an ex vivo porcine heart.

41563_2020_814_MOESM4_ESM.mp4

Supplementary Video 2 In vivo epicardial ECG recording by electrodes with the e-bioadhesive interface on a beating rat heart.

Supplementary Video 3 In vivo stimulation of a rat sciatic nerve by electrodes with the e-bioadhesive interface.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deng, J., Yuk, H., Wu, J. et al. Electrical bioadhesive interface for bioelectronics. Nat. Mater. 20, 229–236 (2021). https://doi.org/10.1038/s41563-020-00814-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41563-020-00814-2

This article is cited by

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