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.

  • Protocol
  • Published:

Activation of mechanoluminescent nanotransducers by focused ultrasound enables light delivery to deep-seated tissue in vivo

Abstract

Light is used extensively in biological and medical research for optogenetic neuromodulation, fluorescence imaging, photoactivatable gene editing and light-based therapies. The major challenge to the in vivo implementation of light-based methods in deep-seated structures of the brain or of internal organs is the limited penetration of photons in biological tissue. The presence of light scattering and absorption has resulted in the development of invasive techniques such as the implantation of optical fibers, the insertion of endoscopes and the surgical removal of overlying tissues to overcome light attenuation and deliver it deep into the body. However, these procedures are highly invasive and make it difficult to reposition and adjust the illuminated area in each animal. Here, we detail a noninvasive approach to deliver light (termed ‘deLight’) in deep tissue via systemically injected mechanoluminescent nanotransducers that can be gated by using focused ultrasound. This approach achieves localized light emission with sub-millimeter resolution and millisecond response times in any vascularized organ of living mice without requiring invasive implantation of light-emitting devices. For example, deLight enables optogenetic neuromodulation in live mice without a craniotomy or brain implants. deLight provides a generalized method for applications that require a light source in deep tissues in vivo, such as deep-brain fluorescence imaging and photoactivatable genome editing. The implementation of the entire protocol for an in vivo application takes ~1–2 weeks.

Key points

  • This protocol describes the synthesis and characterization of mechanoluminescent nanotransducers covering the entire visible spectrum, their characterization in response to ultrasound in tissue-mimicking phantoms and in artificial circulatory systems and their use in mouse behavioral and immunohistochemical assays.

  • Alternative deep-tissue light-delivery approaches include implanted optical fibers or micro light-emitting diodes, intracranially injected upconversion nanoparticles and wavefront-shaping methods based on spatial light modulators.

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

Fig. 1: Principles of deLight and its application in vivo.
Fig. 2: Synthesis of bulk mechanoluminescent materials via solid-state reactions.
Fig. 3: Bright-field and afterglow images of as-synthesized bulk mechanoluminescent materials.
Fig. 4: Synthesis process of biocompatible MLNTs from bulk mechanoluminescent materials.
Fig. 5: Calibration of the FUS system and characterizations of MLNTs in a tissue-mimicking phantom.
Fig. 6: Characterizations of MLNTs in an artificial circulatory system.
Fig. 7: Application of deLight in vivo.
Fig. 8: Dissection and staining of brain sections to evaluate the efficacy of deLight.
Fig. 9: Representative results of in vivo sono-optogenetic neuromodulation with deLight.

Similar content being viewed by others

Data availability

The source data for Extended Data Figs. 38 can be downloaded from https://doi.org/10.6084/m9.figshare.23690961. The STL file of the holder can be found at https://doi.org/10.6084/m9.figshare.23691309. A representative tiff file of the time series mechanoluminescence emission can be found at https://doi.org/10.6084/m9.figshare.23691312. Further data are available from the corresponding author upon request. Characterized and quality-controlled MLNTs are available to other research laboratories upon request.

Code availability

The custom LabVIEW and MATLAB codes used in this protocol are available at https://github.com/ShanJiang1233/deLight and are archived in Zenodo at https://doi.org/10.5281/zenodo.8162191.

References

  1. Prominski, A. et al. Porosity-based heterojunctions enable leadless optoelectronic modulation of tissues. Nat. Mater. 21, 647–655 (2022).

    Article  CAS  PubMed  Google Scholar 

  2. Tian, B. Nongenetic neural control with light. Science 365, 457 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Xu, C. et al. Nanoparticles with ultrasound-induced afterglow luminescence for tumour-specific theranostics. Nat. Biomed. Eng. 7, 298–312 (2023).

    Article  CAS  PubMed  Google Scholar 

  4. Sebesta, C. et al. Subsecond multichannel magnetic control of select neural circuits in freely moving flies. Nat. Mater. 21, 951–958 (2022).

    Article  CAS  PubMed  Google Scholar 

  5. Chen, J. C. et al. A wireless millimetric magnetoelectric implant for the endovascular stimulation of peripheral nerves. Nat. Biomed. Eng. 6, 706–716 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Jiang, S. et al. Spatially expandable fiber-based probes as a multifunctional deep brain interface. Nat. Commun. 11, 6115 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yun, S. H. & Kwok, S. J. J. Light in diagnosis, therapy and surgery. Nat. Biomed. Eng. 1, 0008 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jiang, S., Wu, X., Rommelfanger, N. J., Ou, Z. & Hong, G. Shedding light on neurons: optical approaches for neuromodulation. Natl Sci. Rev. 9, nwac007 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hong, G., Antaris, A. L. & Dai, H. Near-infrared fluorophores for biomedical imaging. Nat. Biomed. Eng. 1, 0010 (2017).

    Article  CAS  Google Scholar 

  10. Zhang, F. et al. Optogenetic interrogation of neural circuits: technology for probing mammalian brain structures. Nat. Protoc. 5, 439–456 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kim, P. et al. In vivo wide-area cellular imaging by side-view endomicroscopy. Nat. Methods 7, 303–305 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Keahey, P., Ramalingam, P., Schmeler, K. & Richards-Kortum, R. R. Differential structured illumination microendoscopy for in vivo imaging of molecular contrast agents. Proc. Natl Acad. Sci. USA 113, 10769–10773 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Yang, Y. et al. Preparation and use of wireless reprogrammable multilateral optogenetic devices for behavioral neuroscience. Nat. Protoc. 17, 1073–1096 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ausra, J. et al. Wireless, battery-free, subdermally implantable platforms for transcranial and long-range optogenetics in freely moving animals. Proc. Natl Acad. Sci. USA 118, e2025775118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Montgomery, K. L. et al. Wirelessly powered, fully internal optogenetics for brain, spinal and peripheral circuits in mice. Nat. Methods 12, 969–974 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chen, S. et al. Near-infrared deep brain stimulation via upconversion nanoparticle-mediated optogenetics. Science 359, 679–684 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Wu, X. et al. Tether-free photothermal deep-brain stimulation in freely behaving mice via wide-field illumination in the near-infrared-II window. Nat. Biomed. Eng. 6, 754–770 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ruan, H. et al. Deep tissue optical focusing and optogenetic modulation with time-reversed ultrasonically encoded light. Sci. Adv. 3, eaao5520 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Hong, G. Seeing the sound. Science 369, 638 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wu, X. et al. Sono-optogenetics facilitated by a circulation-delivered rechargeable light source for minimally invasive optogenetics. Proc. Natl Acad. Sci. USA 116, 26332–26342 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wang, W. et al. Ultrasound-triggered in situ photon emission for noninvasive optogenetics. J. Am. Chem. Soc. 145, 1097–1107 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang, F. et al. Palette of rechargeable mechanoluminescent fluids produced by a biomineral-inspired suppressed dissolution approach. J. Am. Chem. Soc. 144, 18406–18418 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yang, F. et al. A biomineral-inspired approach of synthesizing colloidal persistent phosphors as a multicolor, intravital light source. Sci. Adv. 8, eabo6743 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang, F. et al. Principles and applications of sono-optogenetics. Adv. Drug Deliv. Rev. 194, 114711 (2023).

    Article  CAS  PubMed  Google Scholar 

  25. Zhou, X. X. et al. A single-chain photoswitchable CRISPR-Cas9 architecture for light-inducible gene editing and transcription. ACS Chem. Biol. 13, 443–448 (2018).

    Article  CAS  PubMed  Google Scholar 

  26. Li, X., Lovell, J. F., Yoon, J. & Chen, X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat. Rev. Clin. Oncol. 17, 657–674 (2020).

    Article  PubMed  Google Scholar 

  27. Kim, T.-I. et al. Injectable, cellular-scale optoelectronics with applications for wireless optogenetics. Science 340, 211–216 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Salatino, J. W., Ludwig, K. A., Kozai, T. D. Y. & Purcell, E. K. Glial responses to implanted electrodes in the brain. Nat. Biomed. Eng. 1, 862–877 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Shahriari, D., Rosenfeld, D. & Anikeeva, P. Emerging frontier of peripheral nerve and organ interfaces. Neuron 108, 270–285 (2020).

    Article  CAS  PubMed  Google Scholar 

  30. Hibberd, T. J. et al. Optogenetic induction of colonic motility in mice. Gastroenterology 155, 514–528.e6 (2018).

    Article  PubMed  Google Scholar 

  31. Xian, Q. et al. Modulation of deep neural circuits with sonogenetics. Proc. Natl Acad. Sci. USA 120, e2220575120 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Airan, R. D. & Butts Pauly, K. Hearing out ultrasound neuromodulation. Neuron 98, 875–877 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Sato, T., Shapiro, M. G. & Tsao, D. Y. Ultrasonic neuromodulation causes widespread cortical activation via an indirect auditory mechanism. Neuron 98, 1031–1041.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Guo, H. et al. Ultrasound produces extensive brain activation via a cochlear pathway. Neuron 98, 1020–1030.e4 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Mohammadjavadi, M. et al. Elimination of peripheral auditory pathway activation does not affect motor responses from ultrasound neuromodulation. Brain Stimul. 12, 901–910 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Ye, P. P., Brown, J. R. & Pauly, K. B. Frequency dependence of ultrasound neurostimulation in the mouse brain. Ultrasound Med. Biol. 42, 1512–1530 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Yang, J.-M. et al. Simultaneous functional photoacoustic and ultrasonic endoscopy of internal organs in vivo. Nat. Med. 18, 1297–1302 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Arenkiel, B. R. et al. In vivo light-induced activation of neural circuitry in transgenic mice expressing channelrhodopsin-2. Neuron 54, 205–218 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kefauver, J. M., Ward, A. B. & Patapoutian, A. Discoveries in structure and physiology of mechanically activated ion channels. Nature 587, 567–576 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Oh, S.-J. et al. Ultrasonic neuromodulation via astrocytic TRPA1. Curr. Biol. 29, 3386–3401.e8 (2019).

    Article  CAS  PubMed  Google Scholar 

  41. Hu, Z., Chen, S., Yang, Y., Gong, Y. & Chen, H. An affordable and easy-to-use focused ultrasound device for noninvasive and high precision drug delivery to the mouse brain. IEEE Trans. Biomed. Eng. 69, 2723–2732 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

G.H. acknowledges two awards from the NIH (5R00AG056636-04 and 1R34NS127103-01), an NSF CAREER award (2045120), an NSF EAGER award (2217582), a Rita Allen Foundation Scholars Award, a Beckman Technology Development Grant, a grant from the Focused Ultrasound Foundation, a gift from the Spinal Muscular Atrophy (SMA) Foundation, a gift from the Pinetops Foundation, two seed grants from the Wu Tsai Neurosciences Institute and a seed grant from the Bio-X Initiative of Stanford University. X.W. acknowledges support from a Stanford Graduate Fellowship. N.J.R. acknowledges support from the NSF Graduate Research Fellowships Program (GRFP) and a Stanford Bio-X fellowship. Some schematics were created with BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

S.J., X.W., F.Y., N.J.R. and G.H. contributed ideas and designed research. S.J. and F.Y. synthesized MLNTs. S.J., X.W., F.Y. and N.J.R. characterized MLNTs in PDMS phantoms and artificial circulatory systems under FUS. S.J. and X.W. performed the application of the deLight system for noninvasive sono-optogenetic neuromodulation in live mice. S.J., X.W., F.Y., N.J.R. and G.H. wrote the paper.

Corresponding author

Correspondence to Guosong Hong.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Protocols thanks Hao Shen, Seok-Hyun Yun and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Related links

Key references using this protocol

Hong, G. Science 369, 638 (2020): https://doi.org/10.1126/science.abd3636

Wang, W. et al. J. Am. Chem. Soc. 145, 1097–1107 (2023): https://doi.org/10.1021/jacs.2c10666

Yang, F. et al. J. Am. Chem. Soc. 144, 18406–18418 (2022): https://doi.org/10.1021/jacs.2c06724

Yang, F. et al. Sci. Adv. 8, eabo6743 (2022): https://doi.org/10.1126/sciadv.abo6743

Extended data

Extended Data Fig. 1

Representative TEM images of colloidal MLNTs of Sr2MgSi2O7:Eu,Dy, ZnS:Cu,Al, ZnS:Mn and CaTiO3:Pr. Figure reproduced with permission from ref. 22, ACS.

Extended Data Fig. 2 Peak pressure measured by the hydrophone (black) plotted against the percent amplitude of the FUS system.

Linear fitting (black) yields an empirical formula: P = 0.1A + 0.2, where P is the pressure in mega pascals and A is the amplitude in percent. Figure reproduced from ref. 20. under the Creative Commons license CC BY 4.0.

Extended Data Fig. 3

Pressure mapping of the 1.5-MHz FUS transducer in the x-y plane (at z0 = 0) (a) and in the x-z plane (at y0 = 0) (b).

Extended Data Fig. 4

Normalized mechanoluminescence spectra of MLNTs composed of Sr2MgSi2O7:Eu,Dy (a), ZnS:Cu,Al (b), ZnS:Mn (c) and CaTiO3:Pr (d). Figure adapted with permission from ref. 22, ACS.

Extended Data Fig. 5

An example mechanoluminescence image (a) and its corresponding emission line profile fitted to a Gaussian function (b) to obtain the full-width-at-half-maximum (FWHM).

Extended Data Fig. 6

Representative rechargeability curves of MLNTs composed of Sr2MgSi2O7:Eu,Dy (a), ZnS:Cu,Al (b), ZnS:Mn (c) and CaTiO3:Pr (d). Figure reproduced with permission from ref. 22, ACS.

Extended Data Fig. 7 Temporal kinetics of FUS-induced light emission from colloidal solutions of MLNTs.

a−d, Temporal kinetics of MLNT solutions composed of Sr2MgSi2O7:Eu,Dy (a), ZnS:Cu,Al (b), ZnS:Mn (c) and CaTiO3:Pr (d) upon 10-, 20- and 50-ms FUS pulses in the artificial circulatory system with an imaging frame rate of 200 Hz. e−h, Onset and offset times of mechanoluminescence emission measured from a–d. All data are presented as the mean ± s.d. of 10 independent measurements. ML, mechanoluminescence. Figure reproduced with permission from ref. 22, ACS.

Extended Data Fig. 8

Emission intensity from mechanoluminescent fluids composed of Sr2MgSi2O7:Eu,Dy (a), ZnS:Cu,Al (b), ZnS:Mn (c) and CaTiO3:Pr (d) under varying FUS pressure. Figure reproduced with permission from ref. 22, ACS.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jiang, S., Wu, X., Yang, F. et al. Activation of mechanoluminescent nanotransducers by focused ultrasound enables light delivery to deep-seated tissue in vivo. Nat Protoc 18, 3787–3820 (2023). https://doi.org/10.1038/s41596-023-00895-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-023-00895-8

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