Remote excitation of neuronal circuits using low-intensity, low-frequency ultrasound

William J Tyler, Yusuf Tufail, Michael Finsterwald, Monica L Tauchmann, Emily J Olson, Cassondra Majestic, William J Tyler, Yusuf Tufail, Michael Finsterwald, Monica L Tauchmann, Emily J Olson, Cassondra Majestic

Abstract

Possessing the ability to noninvasively elicit brain circuit activity yields immense experimental and therapeutic power. Most currently employed neurostimulation methods rely on the somewhat invasive use of stimulating electrodes or photon-emitting devices. Due to its ability to noninvasively propagate through bone and other tissues in a focused manner, the implementation of ultrasound (US) represents a compelling alternative approach to current neuromodulation strategies. Here, we investigated the influence of low-intensity, low-frequency ultrasound (LILFU) on neuronal activity. By transmitting US waveforms through hippocampal slice cultures and ex vivo mouse brains, we determined LILFU is capable of remotely and noninvasively exciting neurons and network activity. Our results illustrate that LILFU can stimulate electrical activity in neurons by activating voltage-gated sodium channels, as well as voltage-gated calcium channels. The LILFU-induced changes in neuronal activity were sufficient to trigger SNARE-mediated exocytosis and synaptic transmission in hippocampal circuits. Because LILFU can stimulate electrical activity and calcium signaling in neurons as well as central synaptic transmission we conclude US provides a powerful tool for remotely modulating brain circuit activity.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Generation and propagation of LILFU…
Figure 1. Generation and propagation of LILFU waveforms through neuronal tissue.
(A) General experimental configuration implemented to transmit LILFU waveforms through slice cultures while optically monitoring neuronal activity. (B) Graphical illustration of some of the variables involved in constructing LILFU waveforms. These variables include acoustic frequency (f), the number of acoustic cycles per tone burst (c/tb), tone burst duration (TBD), pulse repetition frequency (PRF), and number of tone bursts per stimulus (Ntb). (C) Acoustic pressure wave (left) produced by a typical US tone burst consisting of 10 acoustic cycles at f = 0.44 MHz and FFT of this US tone burst (right). For the construction of our primary US stimulus waveform (LILFU-1), we used a linearly sweeping PRF by repeating the illustrated tone burst from 0–100 Hz over a 5 sec period.
Figure 2. LILFU stimulates sodium transients mediated…
Figure 2. LILFU stimulates sodium transients mediated by voltage-gated sodium channels in hippocampal neurons.
(A) Confocal image (left) of a slice culture loaded with CoroNa Green AM. Hippocampal regions CA1 stratum pyramidale (SP) and stratum radiatum (SR) are illustrated. Individual (black) and averaged (color) Na+ transients (right) triggered in CA1 pyramidal neuron somas by LILFU-1 under control conditions and in the presence of TTX. (B) Voltage trace of membrane voltage in response to five US tone bursts delivered at a PRF of 10 Hz during whole-cell current clamp recordings of a CA1 pyramidal neuron. (C) Neuronal membrane integrity is preserved following chronic in vitro stimulation with LILFU. Confocal images of CA1 pyramidal neurons from hippocampal slice cultures prepared from thy-1-YFP mice. The images shown are from a control slice culture (left) and a slice culture following chronic stimulation (right) with LILFU-1 every 8 min for 48 h (360 LILFU-1 stimuli). (D) Similar to (C), but higher magnification images of regions in CA1 SR, which more clearly illustrate the presence of fine membrane structures such as dendritic spines for control (top) and chronic LILFU stimulation conditions (bottom).
Figure 3. LILFU triggers voltage-dependent somatic and…
Figure 3. LILFU triggers voltage-dependent somatic and presynaptic Ca2+ transients in neurons.
(A) Confocal image (left) of a slice culture loaded with OGB-1 AM (green) to monitor Ca2+ activity and Sulforhodamine 101 (red) to identify glial cells (yellow). Representative LILFU-triggered Ca2+ transients observed in the somas of neurons and glial cells are illustrated (right). (B) Individual (black) and averaged (green) Ca2+ transients observed in the somas of neurons in response to a brief LILFU waveform. The histogram (inset) illustrates trial 1 normalized mean Ca2+ transient amplitudes in response to repeated trials of LILFU stimulation (n = 19 cells from 3 slices). (C) Confocal image (left) of a slice culture loaded with OGB-1 AM illustrating en passant boutons located in CA1 SR. Individual (black) and averaged (green) presynaptic Ca2+ transients (right) produced by stimulation with LILFU-1. (D) Averaged somatic Ca2+ transients obtained from neurons under control conditions or in the presence of either TTX (n = 36 from 4 slices) or Cd2+ (n = 30 from 4 slices) in response to stimulation with LILFU-1.
Figure 4. LILFU waveforms transmitted through whole…
Figure 4. LILFU waveforms transmitted through whole brains are capable of stimulating calcium transients.
(A) Illustration of basic experimental procedure we developed to transmit LILFU waveforms through whole ex vivo brains prepared from adult wild-type mice and bath-loaded with OGB-1 AM. As depicted, LILFU waveforms were transmitted from the ventral surface of the brain through the tissue to the dorsal surface where we performed confocal imaging. (B) Individual (black) and averaged (green) Ca2+ transients observed in the somas of cells on the dorsal surface of an ex vivo brain in response to stimulation with LILFU-1, which was transmitted through the brain from the ventral surface. (C) Confocal images illustrating OGB-1 loaded cells on the dorsal surface of the brain. The image on left illustrates cells during baseline, while the image on the right illustrates cells two-seconds after stimulation with LILFU-1 ensued.
Figure 5. LILFU stimulates SNARE-mediated synaptic vesicle…
Figure 5. LILFU stimulates SNARE-mediated synaptic vesicle exocytosis and central synaptic transmission.
(A) Confocal images illustrating spH signals obtained before (left) and during (right) stimulation with LILFU-1. (B) Individual (black) and averaged (green) spH signals typically obtained in response to stimulation with LILFU-1. (C) Acoustic pressure wave (left) produced by a single LILFU tone burst consisting of 50,000 acoustic cycles at f = 0.67 MHz and FFT of LILFU tone burst (right). (D) Individual (black) and averaged (green) spH signals obtained in response to stimulation with the LILFU tone burst shown in (C) delivered at a PRF = 10 Hz for 0.5 s to produce Np = 5. (E) Histogram of spH responses obtained as a function of acoustic intensity. Responses from individual experiments are indicated by black crosses while the average response is indicated by the green line. (F) Averaged spH signals illustrating the effect of CNQX+APV (n = 84 from 4 slices), TTX (n = 108 from 4 slices), or BoNT/A (n = 60 from 4 slices) on synaptic vesicle exocytosis induced by LILFU-1. (G) Averaged spH signals obtained from buttons in response to field stimulation of Schaffer collaterals with 250 AP, 50 Hz (n = 48), 100 AP, 20 Hz (n = 63), 40 AP, 20 Hz (n = 51), or by LILFU-1 (n = 148).
Figure 6. Influence of LILFU on putative…
Figure 6. Influence of LILFU on putative excitatory hippocampal CA3-CA1 synapses.
(A) Confocal images illustrating spH expression in CA1 SR (left) and an apical dendritic branch of a CA1 pyramidal neuron, which was labeled with DiI using a DiOlistic labeling technique (middle). The two-channel confocal image (right) illustrates putative excitatory synapses indicated by apposition of spH+ puncta and dendritic spines. (B) Individual (black), mean spH (green), and mean DiI (red) signals obtained from terminals impinging on dendritic spines in response to stimulation with LILFU-1.

References

    1. Wagner T, Valero-Cabre A, Pascual-Leone A. Noninvasive Human Brain Stimulation. Annu Rev Biomed Eng. 2007;9:527–565.
    1. Zhang F, Wang LP, Brauner M, Liewald JF, Kay K, et al. Multimodal fast optical interrogation of neural circuitry. Nature. 2007;446:633–639.
    1. Gavrilov LR, Gersuni GV, Ilyinsky OB, Sirotyuk MG, Tsirulnikov EM, et al. The effect of focused ultrasound on the skin and deep nerve structures of man and animal. Prog Brain Res. 1976;43:279–292.
    1. Fry FJ, Ades HW, Fry WJ. Production of reversible changes in the central nervous system by ultrasound. Science. 1958;127:83–84.
    1. ter Haar G. Therapeutic applications of ultrasound. Prog Biophys Mol Biol. 2007;93:111–129.
    1. Hynynen K, Jolesz FA. Demonstration of potential noninvasive ultrasound brain therapy through an intact skull. Ultrasound Med Biol. 1998;24:275–283.
    1. Clement GT. Perspectives in clinical uses of high-intensity focused ultrasound. Ultrasonics. 2004;42:1087–1093.
    1. Clement GT, Hynynen K. A non-invasive method for focusing ultrasound through the human skull. Phys Med Biol. 2002;47:1219–1236.
    1. Fry WJ. Electrical stimulation of brain localized without probes–theoretical analysis of a proposed method. J Acoust Soc Am. 1968;44:919–931.
    1. Gavrilov LR, Tsirulnikov EM, Davies IA. Application of focused ultrasound for the stimulation of neural structures. Ultrasound Med Biol. 1996;22:179–192.
    1. O'Brien WD., Jr Ultrasound-biophysics mechanisms. Prog Biophys Mol Biol. 2007;93:212–255.
    1. Dinno MA, Dyson M, Young SR, Mortimer AJ, Hart J, et al. The significance of membrane changes in the safe and effective use of therapeutic and diagnostic ultrasound. Phys Med Biol. 1989;34:1543–1552.
    1. Dalecki D. Mechanical bioeffects of ultrasound. Annu Rev Biomed Eng. 2004;6:229–248.
    1. Mortimer AJ, Dyson M. The effect of therapeutic ultrasound on calcium uptake in fibroblasts. Ultrasound Med Biol. 1988;14:499–506.
    1. Chapman IV, MacNally NA, Tucker S. Ultrasound-induced changes in rates of influx and efflux of potassium ions in rat thymocytes in vitro. Ultrasound Med Biol. 1980;6:47–58.
    1. Sukharev S, Corey DP. Mechanosensitive channels: multiplicity of families and gating paradigms. Sci STKE. 2004;2004:re4.
    1. Morris CE, Juranka PF. Lipid Stress at Play: Mechanosensitivity of Voltage-Gated Channels. Academic Press; 2007. pp. 297–338. Current Topics in Membranes:
    1. Mihran RT, Barnes FS, Wachtel H. Temporally-specific modification of myelinated axon excitability in vitro following a single ultrasound pulse. Ultrasound Med Biol. 1990;16:297–309.
    1. Tsui PH, Wang SH, Huang CC. In vitro effects of ultrasound with different energies on the conduction properties of neural tissue. Ultrasonics. 2005;43:560–565.
    1. Rinaldi PC, Jones JP, Reines F, Price LR. Modification by focused ultrasound pulses of electrically evoked responses from an in vitro hippocampal preparation. Brain Res. 1991;558:36–42.
    1. Bachtold MR, Rinaldi PC, Jones JP, Reines F, Price LR. Focused ultrasound modifications of neural circuit activity in a mammalian brain. Ultrasound Med Biol. 1998;24:557–565.
    1. Foster KR, Wiederhold ML. Auditory responses in cats produced by pulsed ultrasound. J Acoust Soc Am. 1978;63:1199–1205.
    1. Young RR, Henneman E. Functional effects of focused ultrasound on mammalian nerves. Science. 1961;134:1521–1522.
    1. Fry WJ, Wulff VJ, Tucker D, Fry FJ. Physical factors involved in ultrasonically induced changes in living systems: I. Identification of non-temperature effects. J Acoust Soc Am. 1950;22:867–876.
    1. White PJ, Clement GT, Hynynen K. Local frequency dependence in transcranial ultrasound transmission. Phys Med Biol. 2006;51:2293–2305.
    1. Hayner M, Hynynen K. Numerical analysis of ultrasonic transmission and absorption of oblique plane waves through the human skull. J Acoust Soc Am. 2001;110:3319–3330.
    1. Meier SD, Kovalchuk Y, Rose CR. Properties of the new fluorescent Na+ indicator CoroNa Green: comparison with SBFI and confocal Na+ imaging. J Neurosci Methods. 2006;155:251–259.
    1. Miller DL, Pislaru SV, Greenleaf JE. Sonoporation: mechanical DNA delivery by ultrasonic cavitation. Somat Cell Mol Genet. 2002;27:115–134.
    1. Feng G, Mellor RH, Bernstein M, Keller-Peck C, Nguyen QT, et al. Imaging neuronal subsets in transgenic mice expressing multiple spectral variants of GFP. Neuron. 2000;28:41–51.
    1. Trevelyan AJ, Sussillo D, Watson BO, Yuste R. Modular propagation of epileptiform activity: evidence for an inhibitory veto in neocortex. J Neurosci. 2006;26:12447–12455.
    1. Paoletti P, Ascher P. Mechanosensitivity of NMDA receptors in cultured mouse central neurons. Neuron. 1994;13:645–655.
    1. Maroto R, Raso A, Wood TG, Kurosky A, Martinac B, et al. TRPC1 forms the stretch-activated cation channel in vertebrate cells. Nat Cell Biol. 2005;7:179–185.
    1. Li Z, Burrone J, Tyler WJ, Hartman KN, Albeanu DF, et al. Synaptic vesicle recycling studied in transgenic mice expressing synaptopHluorin. Proc Natl Acad Sci U S A. 2005;102:6131–6136.
    1. Miesenbock G, De Angelis DA, Rothman JE. Visualizing secretion and synaptic transmission with pH-sensitive green fluorescence proteins. Nature. 1998;394:192–195.
    1. Gan WB, Grutzendler J, Wong WT, Wong RO, Lichtman JW. Multicolor “DiOlistic” labeling of the nervous system using lipophilic dye combinations. Neuron. 2000;27:219–225.
    1. Rosenmund C, Stevens CF. Definition of the readily releasable pool of vesicles at hippocampal synapses. Neuron. 1996;16:1197–1207.
    1. Bronk P, Deak F, Wilson MC, Liu X, Sudhof TC, et al. Differential effects of SNAP-25 deletion on Ca2+ -dependent and Ca2+ -independent neurotransmission. J Neurophysiol. 2007;98:794–806.
    1. Sankaranarayanan S, De Angelis D, Rothman JE, Ryan TA. The use of pHluorins for optical measurements of presynaptic activity. Biophys J. 2000;79:2199–2208.
    1. Burrone J, Li Z, Murthy VN. Studying vesicle cycling in presynaptic terminals using the genetically encoded probe synaptopHluorin. Nat Protoc. 2006;1:2970–2978.
    1. Ang ES, Jr, Gluncic V, Duque A, Schafer ME, Rakic P. Prenatal exposure to ultrasound waves impacts neuronal migration in mice. Proc Natl Acad Sci U S A. 2006;103:12903–12910.
    1. Ebisawa K, Hata K, Okada K, Kimata K, Ueda M, et al. Ultrasound enhances transforming growth factor beta-mediated chondrocyte differentiation of human mesenchymal stem cells. Tissue Eng. 2004;10:921–929.
    1. Reher P, Doan N, Bradnock B, Meghji S, Harris M. Effect of ultrasound on the production of IL-8, basic FGF and VEGF. Cytokine. 1999;11:416–423.
    1. Mukai S, Ito H, Nakagawa Y, Akiyama H, Miyamoto M, et al. Transforming growth factor-beta1 mediates the effects of low-intensity pulsed ultrasound in chondrocytes. Ultrasound Med Biol. 2005;31:1713–1721.
    1. Naruse K, Miyauchi A, Itoman M, Mikuni-Takagaki Y. Distinct anabolic response of osteoblast to low-intensity pulsed ultrasound. J Bone Miner Res. 2003;18:360–369.
    1. Sant'Anna EF, Leven RM, Virdi AS, Sumner DR. Effect of low intensity pulsed ultrasound and BMP-2 on rat bone marrow stromal cell gene expression. J Orthop Res. 2005;23:646–652.
    1. Abe K, Saito H. Effects of basic fibroblast growth factor on central nervous system functions. Pharmacol Res. 2001;43:307–312.
    1. Molteni R, Fumagalli F, Magnaghi V, Roceri M, Gennarelli M, et al. Modulation of fibroblast growth factor-2 by stress and corticosteroids: from developmental events to adult brain plasticity. Brain Res Brain Res Rev. 2001;37:249–258.
    1. Charron F, Tessier-Lavigne M. The Hedgehog, TGF-beta/BMP and Wnt families of morphogens in axon guidance. Adv Exp Med Biol. 2007;621:116–133.
    1. Gora-Kupilas K, Josko J. The neuroprotective function of vascular endothelial growth factor (VEGF). Folia Neuropathol. 2005;43:31–39.
    1. Jin KL, Mao XO, Greenberg DA. Vascular endothelial growth factor: direct neuroprotective effect in in vitro ischemia. Proc Natl Acad Sci U S A. 2000;97:10242–10247.
    1. Flanders KC, Ren RF, Lippa CF. Transforming growth factor-betas in neurodegenerative disease. Prog Neurobiol. 1998;54:71–85.
    1. Tesseur I, Wyss-Coray T. A role for TGF-beta signaling in neurodegeneration: evidence from genetically engineered models. Curr Alzheimer Res. 2006;3:505–513.
    1. Mattson MP. NF-kappaB in the survival and plasticity of neurons. Neurochem Res. 2005;30:883–893.
    1. Guo W, Jiang H, Gray V, Dedhar S, Rao Y. Role of the integrin-linked kinase (ILK) in determining neuronal polarity. Dev Biol. 2007;306:457–468.
    1. Brunet A, Datta SR, Greenberg ME. Transcription-dependent and -independent control of neuronal survival by the PI3K-Akt signaling pathway. Curr Opin Neurobiol. 2001;11:297–305.
    1. Hsu HC, Fong YC, Chang CS, Hsu CJ, Hsu SF, et al. Ultrasound induces cyclooxygenase-2 expression through integrin, integrin-linked kinase, Akt, NF-kappaB and p300 pathway in human chondrocytes. Cell Signal. 2007;19:2317–2328.
    1. Tang CH, Yang RS, Huang TH, Lu DY, Chuang WJ, et al. Ultrasound stimulates cyclooxygenase-2 expression and increases bone formation through integrin, focal adhesion kinase, phosphatidylinositol 3-kinase, and Akt pathway in osteoblasts. Mol Pharmacol. 2006;69:2047–2057.
    1. Magee TR, Davies AH. Auditory phenomena during transcranial Doppler insonation of the basilar artery. J Ultrasound Med. 1993;12:747–750.
    1. Hynynen K, Clement GT, McDannold N, Vykhodtseva N, King R, et al. 500-element ultrasound phased array system for noninvasive focal surgery of the brain: a preliminary rabbit study with ex vivo human skulls. Magn Reson Med. 2004;52:100–107.
    1. Andrews RJ. Neuroprotection trek–the next generation: neuromodulation I. Techniques–deep brain stimulation, vagus nerve stimulation, and transcranial magnetic stimulation. Ann N Y Acad Sci. 2003;993:1–13; discussion 48-53.
    1. Stoppini L, Buchs PA, Muller D. A simple method for organotypic cultures of nervous tissue. J Neurosci Methods. 1991;37:173–182.
    1. NEMA . Acoustic Output Measurement Standard for Diagnostic Ultrasound Equipment. Washington, DC: National Electrical Manufacturers Association; 2004.

Source: PubMed

3
Sottoscrivi