Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

RESEARCH ARTICLE

Neuromodulation of Chemical Synaptic


Transmission Driven by THz Photons Citation: Tan X, Zhong Y, Li R,
Xiaoxuan Tan1,2*, Yuan Zhong1,3, Ruijie Li4, and Chao Chang1,3* Chang C. Neuromodulation of
Chemical Synaptic Transmission
1
Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Driven by THz Photons. Research
2022;2022:Article 0010. https://doi.
Beijing 100071, China. 2Astronaut Center of China, Beijing 100084, China. 3Department of Engineering org/10.34133/research.0010
Physics, Tsinghua University, Beijing 100084, China. 4Brain Research Center and State Key Laboratory
Submitted 10 July 2022
of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing 400038, China. Accepted 25 October 2022
Published 19 December 2022
*Address correspondence to: t15822082707@163.com (X.T.); changc@xjtu.edu.cn (C.C.)
Copyright © 2022 Xiaoxuan Tan
et al. Exclusive Licensee Science and
Postsynaptic currents of chemical synapse are modulated by multitudinous neurotransmitters, such Technology Review Publishing House.
as acetylcholine, dopamine, glutamate, and γ-aminobutyric acid, many of which have been used in the No claim to original U.S. Government
treatment of neurological diseases. Here, based on molecular dynamics simulations and quantum chemical Works. Distributed under a Creative
calculation, we propose that 30- to 45-THz photons can resonate with a variety of typical neurotransmitter Commons Attribution License

Downloaded from https://spj.science.org on April 06, 2023


molecules and make them absorb photon energy to activate the transition to high energy state, which is (CC BY 4.0).
expected to be a new method of neural regulation. Furthermore, we verified the calculated results through
experiments that THz irradiation could substantively change neuronal signal emission and enhance
the frequency, amplitude, and dynamic properties of excitatory postsynaptic current and inhibitory
postsynaptic current. In addition, we demonstrated the potential of neural information regulation by THz
photons through 2-photon imaging in vivo. These findings are expected to improve the understanding of
the physical mechanism of biological phenomena and facilitate the application of terahertz technology
in neural regulation and the development of new functional materials.

Introduction ultraviolet radiation has not observed similar effects [9]. This
suggests that the regulation of the neural information by the
Since the 18th century, paradigms used to describe the nature terahertz wave is a thermal effect, but the specific THz photons
of light and its interaction with matter have gone through may be dominant in the physiological process.
Newton’s theory of light particles, Maxwell’s wave paradigm According to the traditional viewpoint of molecular phys-
of light, and Planck and Einstein’s theory of photons, until de ics, biophoton is generated during the transition of living mat-
Broglie unified matter and light and proposed the wave-­particle ter from high energy state in the process of biological system
duality of light. When there is no strong interaction between metabolism. However, due to the extremely weak energy of
photons and matter, photons behave like a wave, and when it biophoton radiation and the lack of sensitive detection meth-
has a strong interaction with matter (absorption or emission), ods, the relevant mechanism and significance have not been
photons act more like particles [1–3]. clarified [10–14]. The concept of a photon as a particle also
Along come more and more scientists who believed that leads many researchers to use it as a “massless reagent,” which
consciousness was a matter [4]. The generation, transmission, can react with molecules and be absorbed after collision, lead-
and coupling of neural signals should conform to electromag- ing to a reaction of R + hv → *R, and the number of photons
netic fields and quantum theory. The quantum energy states can be calculated according to E = N0hv = N0(c/λ) [15]. In
of ions and molecules in the nervous system are important order to study the interaction between terahertz waves and biol­­
factors for signal generation, amplification, and coupling. The ogical systems, a large number of studies have been reported,
storage of biological information should be based on molec- confirming that the interaction between terahertz waves and
ular quantum states [5,6]. There are also those who believe that various substances is universal, and it can affect the physical
consciousness has nothing to do with quantum mechanics. and chemical properties of both organic and inorganic mate-
They argue that the brain is a wet, warm system in which deco- rials. Terahertz is important for many biotechnology- and
herence destroys the quantum superpositions of neuron firing engineering-related phenomena, such as simulation studies
much faster than we thought, preventing our brains from act- on the influence of terahertz on water filtration [16], DNA
ing as quantum computers [7]. However, recent studies have unwinding [17,18], ion channel permeability [19], and myelin
shown that terahertz wave irradiation in the low absorption sheath information transduction [20–22]. For example, recent
band of water can significantly change the waveform and emis- theoretical studies revealed that THz waves can promote the
sion frequency of neuronal action potential (AP) [8], and unwinding of DNA due to the resonance of THz waves with
effectively improve the learning ability of animals ~50%, while the vibration of DNA bases [17], and it can also induce structural

Tan et al. 2022 | https://doi.org/10.34133/research.0010 1


Results and Discussion

The ground state spectra of neurotransmitters


Infrared spectra and Raman spectra are often combined to
obtain molecular vibration information. When polar groups
and asymmetric molecules absorb infrared radiation during the
vibration process, the dipole moments change, and the molecule
transitions from the ground state to the excited state, resulting
in the infrared absorption spectrum. In addition, the external
electric field of the incident photon will cause the deformation
of the electron shell of the molecule, and the relative movement
of the positive and negative charge centers of the molecule will
form the induced dipole moment, resulting in polarization phe-
nomenon, which is the formation principle of Raman spectrum
[30–33]. In density functional (DFT) calculation, we use a har-
monic oscillator model to calculate the vibration frequency and
calculate the second derivative of energy with respect to the coor-
Fig. 1. (A) Synaptic connection diagram. (B) Molecular structure of glutamate and
dinate to get the corresponding resonance potential function.
GABA. (C) THz can stimulate the neurotransmitter to release photons. (D) THz
The various functional neural networks formed by synaptic
photons enhance postsynaptic currents in  vitro. (E) THz photons alter neuronal
excitability in vivo (in the actual experiment, the brain of the mice was cut open).
connections between neurons are mainly maintained by a bal-
The blue and red traces in (D) and (E) represent synaptic and Ca2+ signals from ance between glutamate-mediated excitation and GABA-mediated
recorded neurons. inhibition [21–24]. Therefore, we calculated the spectra of gluta-

Downloaded from https://spj.science.org on April 06, 2023


mate and GABA. Previous studies [8] have shown that water
has strong absorption in parts of the terahertz band, mean­­ing that
careful wavelength selection is critical for neurotransmitter
transition and superpermeation of confined monolayer water regulation (Fig. 2A). As shown in Fig. 2B, both neurotransmitters
[16]. Besides, there were also studies that proposed that myelin have strong resonance absorption peaks at 30 to 45 THz, while
can serve as an infrared dielectric waveguide because the mye- water has very weak absorption of THz wave at this frequency
lin sheath possesses a ≈2-fold higher refraction index com- band, so 30 to 45 THz can act directly on the target molecule
pared to the outer medium or the inner axon with a certain without being absorbed by the abundant water in the biological
mid-infrared to terahertz spectral range. Nevertheless, the quan­­ system. In addition, according to Raman spectra (Fig. 2C), the
tum mechanisms underlying their molecular dynamics and polarization changes of the 2 neurotransmitters caused by 30 to
chemical reactivity are not fully understood. At the same time, 45 THz are very small, indicating that the physical principle of
the unique optical, electronic, and biological properties of the activation by THz photons can be attributed to the characteristic
different frequencies of terahertz also need to be translated vibration between electromagnetic wave and neurotransmitter,
into practice in experiments. and the neurotransmitter absorbs radiation and produces elec-
As we know, in addition to the AP that has been shown to tronic state transition.
be mediated by THz photons, communication between neu-
rons, known as synaptic transmission, also plays an important
role in neural signal processing and transmission [23–28], and Neurotransmitter excitation process
the intensity and precision of the quantum postsynaptic poten- Neurotransmitters respond well to terahertz waves at specific
tial are very important to satisfy the conduction velocity of frequencies, including strong absorption of 30 to 45 THz with-
neuron signals [29]. Activation and inactivation of neurotrans- out water interference and subsequent molecular relaxation,
mitters directly affect postsynaptic currents of chemical synapse, encouraging further investigation of the mechanisms involved.
so neurotransmitters provide an excellent option to explore the First, by using DFT and TDDFT calculations, we calculate the
photonic properties of neural information. Here, we calculated geometric structures in the ground state (S0) and the first excited
the properties (infrared and Raman spectra, molecular orbitals, state (S1) to decipher the different conformations of the mol-
excitation energy, fluorescence emission energy, etc.) of 2 typical ecule under study. (Fig. 3A and C and Figs. S7 and S8).
neurotransmitters [glutamate and γ-aminobutyric acid (GABA)] The molecular orbitals involved in the S0–S1 transitions were
via density functional theory (DFT) and time-dependent den- also calculated. In orbitals of glutamate, the highest occupied
sity functional theory (TDDFT) and found the terahertz band molecular orbital (HOMO) and the lowest unoccupied molec-
with characteristic response. Based on this, we used the inde- ular orbital (LUMO) were all spread on the -NH3 group. In
pendently developed quantum cascade THz irradiation system, orbitals of GABA, the HOMO was spread on the -NH3 group,
confirming that THz photons in the corresponding frequency while the LUMO was primarily spread on the -COOH group.
band can significantly change the excitability of neurons and The HOMO energy increased, while the LUMO energy decreased
improve the transmission efficiency of nerve signals through with the strong absorption of THz photons, resulting in a sig-
the patch-clamp and 2-photon imaging in vivo technology. As nificant decrease in the HOMO–LUMO gap. As a result, the
shown in Fig. 1A to E, these results provide conclusive evidence HOMO–LUMO gap of glutamate decreased from 7.57 eV to
that photons are indeed one of the key forms of biological in­­ 4.29 eV, and the HOMO–LUMO gap of GABA decreased from
formation transmission and indicate that terahertz wave mod- 7.31 eV to 5.09 eV (Fig. 3B and D). Thus, the small HOMO–
ulation (THM) has great application potential in the field of LUMO energy gap promotes subsequent molecular relaxation
biological information regulation. and photon liberation. Meanwhile, vertical excitation energy

Tan et al. 2022 | https://doi.org/10.34133/research.0010 2


to 4.47 ± 0.73 ms; n = 12 cells; t11 = 2.985; P = 0.012, paired
Student’s t test), and half-width (from 1.79 ± 0.29 to 1.97 ± 0.39 ms;
n = 11 cells; t10 = 1.761; P = 0.11, paired Student’s t test)
showed no significant change during THM, although the time
to rise by 50% increased (from 0.61 ± 0.04 to 0.67 ± 0.07 ms;
n = 11 cells; t10 = 3.320; P = 0.77 × 10−2, paired Student’s t test;
Fig. 4C and Fig. S9).
Then, we conducted a new set of experiments and tested the
effect of the same power (10 mW) of THz and Blu-ray on the
EPSC separately. As shown in Fig. S11, Blu-ray irradiation also
enhanced the frequency of EPSC (~2.08 times; n = 8 cells; t7 =
4.697; P = 0.0022, paired Student’s t test), but the effect was far
less obvious than THz (~5.51 times; n = 6 cells; t5 = 4.390; P =
0.0071, paired Student’s t test). Besides, THz significantly changed
the amplitude, slope, and area of EPSC, but these parameters
did not change significantly under blue light irradiation, indi-
Fig.  2.  Mechanism by which terahertz enables neurotransmitters to transition to cating that there was a nonthermal effect of EPSC in response
an excited state. (A) Diagram of synaptic connections. (B) Infrared spectra of 2 to THz and the effect produced by THz photons is specific,
neurotransmitters and water. (C) Raman spectra of 2 neurotransmitters. The orange although it could not rule out a thermal effect. We also tested
curves represent the excitatory neurotransmitter glutamate, and the gray curves the effect of THz on evoked currents, which was consistent with
represent the inhibitory neurotransmitter GABA. a.u., arbitrary units. the amplitude change of spontaneous postsynaptic currents,
increasing from 77 ± 4.09 to 147 ± 12.05 pA (n = 6 cells; t5 =
6.452; P =0.0013, paired Student’s t test; Fig. S12).

Downloaded from https://spj.science.org on April 06, 2023


Next, we examined the effects of THz on inhibitory postsyn-
and fluorescence-emitting energy of 2 neurotransmitters were aptic currents (IPSCs). In agreement with calculated results,
calculated (Fig. 3E). THz photons cause many significant changes in IPSC (Fig. 5A
Excitingly, the excited states S1 of glutamate and GABA also and B). The frequency and peak amplitude increased by ~209%
have strong absorption (Figs. S1 to S4) at 30 to 45 THz, so THz (from 8.87 ± 2.81 to 27.44 ± 8.27 Hz; n = 10 cells; P <10−4,
photons are promising for simultaneous neurotransmitter reg- paired Student’s t test) and ~54% (from 15.89 ± 1.89 to 24.47 ±
ulation and rapid detection at the femtosecond scale by pulse- 3.59 pA; n = 10 cells; P <10−4, paired Student’s t test), respec-
probe flash spectroscopy [34]. tively; the 10% to 90% slope of the IPSC rising phase increased
from 11.75 ± 2.61 to 34.83 ± 6.78 pA·ms (n = 10 cells; P <10−4,
THz photons enhances postsynaptic currents in vitro paired Student’s t test), and the area decreased from 86.92 ±
We used an independently designed and manufactured pulsed 15.91 to 65.74 ± 14.84 pA·ms (n = 10 cells; P = 0.9 × 10−3, paired
quantum cascade laser as the THz light source in this study (see Student’s t test). Besides, other dynamic properties of IPSC such
the Supplementary Materials for details). A THz fiber (core as rise 50% time (from 0.77 ± 0.09 to 0.65 ± 0.05 ms; n = 10
diameter, 240 μm; numerical aperture, 0.27) delivered the THz cells; t9 = 4.478; P = 0.15 × 10−2, paired Student’s t test) and
photons to the target region of the brain slice. We delivered the half-width (from 2.81 ± 0.56 to 1.92 ± 0.33 ms; n = 10 cells; t9 =
THz with the following parameters: average irradiation power, 6.002; P = 0.2 × 10−3, paired Student’s t test) also changed sig-
~30 mW; pulse width, 2 μs; repetition rate, 200 kHz. nificantly during THz application (Fig. S10). Similar to EPSC,
To examine the effect of THz on postsynaptic currents, we these changes of IPSC were also reversible and reproducible.
performed whole-cell voltage patch-clamp recording from However, some of the dynamic properties of IPSC show oppo-
layer 5 pyramidal neurons (PNs) in acute slices of mouse pre- site changes to those of EPSC, such as rise 50% time, half-width,
frontal cortex (Fig. 4B and Fig. S16). Close examination of the and area (Fig. 5C and Fig. S10). We speculate that this may be
excitatory postsynaptic current (EPSC) revealed a dramatic due to the different types and numbers of specific bonds that
change in frequency and amplitude (Fig. 4A and B) and the resonate with different neurotransmitters caused by 30- to
change was readily reversible (Fig. 4C). The EPSC frequency 45-THz photons, and the change of vibration mode and fre-
substantially increased by ~64%, from 9.53 ± 1.19 to 15.65 ± quency is also different, which is manifested at the cellular level
1.39 Hz (n = 11 cells; t10 = 4.956; P = 0.6 × 10−3, paired by the different dynamic properties of postsynaptic currents. By
Student’s t test), and the peak amplitude increased by ~46%, recording miniature EPSC (mEPSC) and miniature IPSC (mIPSC),
from 9.75 ± 0.31 to 12.12 ± 1.55 pA (n = 12 cells; t11 = 4.681; we also found that THz photons changed the presynaptic quan-
P = 0.7 × 10−3, paired Student’s t test). We also counted the tal release changes (Figs. S13 and 14). These experiments above
changes of EPSC frequency in different irradiation periods together establish a basic proof of principle for THz photons
and found that the effect of THz on EPSC frequency gradually in vitro, demonstrating that THz energy delivery indeed induces
weakened with time (Fig. S15). In addition, THM significantly neurotransmitter activation in the absence of any exogenous
changed the kinetic properties of EPSC, and the area (from materials.
49.64 ± 4.45 to 56.79 ± 8.15 pA·ms; n = 11 cells; P = 0.72 × 10−2,
paired Student’s t test) and slope (from 6.11 ± 0.67 to 8.03 ±
1.96 pA·ms; n = 11 cells; t10 = 3.602; P = 0.48 × 10−2, paired THz photons alters neuronal excitability in vivo
Student’s t test) were increased during THM. However, the rise Motivated by the above results, we tested whether THz photons
time (from 2.52 ± 0.11 to 2.46 ± 0.16 ms; n = 12 cells; t11 = 1.857; affects neural signals in vivo. As described in a previous study
P = 0.09, paired Student’s t test), decay time (from 4.99 ± 0.72 [9], we first placed the fiber tip closely above the cortical surface

Tan et al. 2022 | https://doi.org/10.34133/research.0010 3


Downloaded from https://spj.science.org on April 06, 2023
Fig. 3. THz activates neurotransmitters and the mechanism illustrations. (A) DFT and TDDFT optimized structures for the ground and excited states of glutamate and (B)
corresponding molecular orbital energies. (C) DFT and TDDFT optimized structures for the ground and excited states of GABA and (D) corresponding molecular orbital energies.
(E) Vertical excitation energy and fluorescence emission energy of glutamate and GABA.

following craniotomy, and the irradiation time was 60 s (Fig. 6A). increased and some decreased during THz application (Fig.
After THz delivery, we performed immunohistochemistry for 6E). Interestingly, within the same cell type, the pattern of change
c-Fos, a widely used molecular marker of neuronal activation. seemed to be consistent, with cells that were excited being
As shown in Fig. 6B, a large number of neurons in the target suppressed and cells that were not excited being activated.
area were activated, and the c-Fos–positive cells can reach layer Out of a total of 270 neurons from 5 animals, we found 116
5 of the cortex. THz-activated neurons (from 0.62 ± 0.08 to 1.64 ± 0.12 per
We next employed 2-photon Ca2+ imaging to visualize live minute; n = 116 cells; t115 = 15.57; P <10−4, paired Student’s
neuronal population activities during THz application (Fig. t test) and 58 THz-suppressed neurons (from 3.89 ± 2.37 to 1.69 ±
6C). Sensory cortex neurons were labeled with a genetically 1.52 per minute; n = 58 cells; t57 = 12.11; P <10−4, paired
encoded Ca2+ indicators, AAV-GCamp6f. After virus expres- Student’s t test; Fig. 6F). The activity level for the THz-activated
sion, we performed real-time (30 Hz) 2-photon imaging over neurons recovered after the THz window, returning to the
the entire course of THz application (2 min before, 4-min THz same level as before irradiation (from 0.62 ± 0.08 to 0.76 ±
irradiation, 2 min after; Fig. 6D). An example animal showed 0.09 per minute; n = 116 cells; t115 = 1.963; P = 0.052, paired
that THz application reliably changed Ca2+ transients during Student’s t test; Fig. 6G), and at the same time, although the
the application time window over different cells. Similar to excitation level of the inhibited cells recovered after irradiation
previous findings that THz photons enhance both EPSC and (from 1.69 ± 1.52 to 2.18 ± 2.30 per minute; n = 58 cells; t57 =
IPSC, imaging results showed that the excitability of some cells 2.280; P = 0.026, paired Student’s t test; Fig. 6H), it did not

Tan et al. 2022 | https://doi.org/10.34133/research.0010 4


Fig. 4. THz enhanced the EPSC. (A) Typical response during terahertz irradiation (shaded in orange). (B) Left: Physical image of THz and whole-cell recording from
a PN in prefrontal cortex (PFC) slice. Right: Example EPSC responses to THz. Gray, control (Ctrl); orange, THz. (C) Group data showing changes in frequency, amplitude,

Downloaded from https://spj.science.org on April 06, 2023


slope, rise 50% time, half-width, and area upon THz. *P < 0.05, **P < 0.01, and ***P < 0.001. Paired Student’s t test and repeated-measures one-way ANOVA. Error bars
represent SEM. n.s., not significant.

Fig. 5. THz enhanced the IPSC. (A) Typical response during terahertz irradiation (shaded in orange). (B) Example IPSC responses to THz. Gray, control (Ctrl); orange, THz. (C)
Group data showing changes in frequency, amplitude, slope, rise 50% time, half-width, and area upon THz. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. Paired
Student’s t test and repeated-measures one-way ANOVA. Error bars represent SEM.

reach the pre-irradiation level (from 3.89 ± 2.37 to 2.18 ± 2.30 com­­plex than in vivo. Nevertheless, our results from patch-
per minute; n = 58 cells; t57 = 7.245; P <10−4, paired Student’s clamp, c-Fos imaging, and 2-photon Ca2+ imaging are highly
t test; Fig. 6H). Note that the proportion of THz-changed neu- consistent in demonstrating that THz photons can change the
rons detected in these 2-photon Ca2+ imaging experiments activity of a substantial number of neurons in the targeted
was 64.44% (including 42.96% activated neurons and 21.48% cortical region.
suppressed neurons), which was lower than that in the patch- It is important to point out that although THM is similar
clamp results (EPSC or IPSC increased in all tested cells). We to the well-known and widely deployed photodynamic ther-
speculate that this difference could result from the differences apy, it can reliably induce photon-triggered molecular re­­
in test subjects, i.e., the synaptic connections between a large sponses in a precisely targeted and spatially confined volume
number of neurons are cut in the brain slice experiment, and the of biological tissue in vivo, and the 2 are fundamentally differ-
neural circuits or interactions between neurons are far less ent. Bio­­compatibility of nanofluorophores is an important

Tan et al. 2022 | https://doi.org/10.34133/research.0010 5


Downloaded from https://spj.science.org on April 06, 2023

Fig. 6. THz photons alter neuronal excitability in vivo. (A) General scheme for targeted THz photon delivery to mouse brain in vivo. (B) Left: Confocal image of a postmortem
slice near the THz target spot. Right: Magnified view of the left image (c-Fos is a commonly used marker of neuronal activation). (C) Cartoon showing the general scheme for
the in vivo 2-photon imaging experiments. (D) An example in vivo 2-photon image (averaged 100 frames). (E) Ca2+ activity traces of 12 example neurons. Excited neurons were
suppressed (left) and neurons that were not excited were activated (right). (F) Proportion of neurons with 3 patterns of change, 270 neurons from 5 animals. (G) Changes in
frequency of Ca2+ signaling in inhibited cells. (H) Changes in frequency of Ca2+ signaling in excited cells. *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. Paired
Student’s t test and repeated-measures one-way ANOVA. Error bars represent SEM.

concern for their in vivo application, but THM does not Conclusion
require injection of exogenous materials, and its effect depends
entirely on the quantity and frequency of photons rather than In this study, we found that THz photons can modulate post-
the properties of encapsulated molecules. It has good stability synaptic currents. It increases the frequency, amplitude, and
and excellent biocompatibility, which facilitate clinical trans- kinetic properties of EPSC and IPSC, which is probably
lation [35–37]. be­c ause THz photons can resonate with a variety of typical

Tan et al. 2022 | https://doi.org/10.34133/research.0010 6


neurotransmitter molecules and make them absorb photon Center in Tianjin. Author contributions: X.T. performed quan-
energy to activate the transition to high energy state. In addi- tum chemical calculations, electrophysiological recordings, and
tion, THz also increases the amplitude of mEPSC, mIPSC, and data analyses. Y.Z. and R.L. contributed to the construction of
evoked currents. Control experiments with Blu-ray stimulation the experimental system. C.C. designed the study. X.T. and C.C.
revealed that THz photons provide nonthermal modulation wrote the manuscript. Competing interests: The authors declare
of chemical synaptic transmission. We further demonstrate that that they have no competing interests.
THz photons could change neuronal excitability. Therefore,
THz photons with a specific wavelength could be a new method Data Availability
to modulate neuronal excitability and synaptic transmission
directly in vitro and in vivo. Data are available from the corresponding author upon request.
On the one hand, our results illustrate that THz photons
could be used as a promising form of brain function modula- Supplementary Materials
tion and brain disease therapy. Most previous studies explore Supplementary Materials and Methods
the method of modulating brain activities by optogenetics, Figs. S1 to S16
which is currently one of the best methods to modulate syn- Tables S1 to S6
aptic transmission. However, the optogenetics technique needs References
to introduce exogenous genes in the brain, and it shows little
potential for applications in healthy humans. Our energy stim-
ulation technique in this study is fundamentally different from References
the optogenetics because it does not require any introduction 1. Jisha CP, Alberucci A, Lee R-K, Assanto G. Optical solitons
of a light-sensitive opsin into the neuronal cells. and wave-particle duality. Opt Lett. 2011;36(10):1848–1850.
On the other hand, our results provide a new theoretical 2. Biktasheva IV, Biktashev VN. Wave-particle dualism of spiral
approach to neuroscience research. There are many phenom-

Downloaded from https://spj.science.org on April 06, 2023


waves dynamics. Phys Rev E. 2003;67(2):026221.
ena that theories of traditional neuroscience cannot explain, 3. Greulich KO. Single molecule experiments challenge the strict
such as why nerve signals travel so fast in myelinated nerves. wave-particle dualism of light. Int J Mol Sci. 2010;11(1):304–311.
A simple flow of charged ions cannot keep up with that speed, 4. Tegmark M. Consciousness as a state of matter. Chaos Soliton
while photons travel extremely fast. One study has established Fract. 2015;76:238–270.
a physical model of neural high-frequency signal generation 5. Liu GZ. The conjectures on physical mechanism of vertebrate
and transmission by quantum optics and electrodynamics nervous system. Chin Sci Bull. 2018;63(36):3864–3865.
methods [21,22]. Our results in this study provide experimental 6. Li N, Peng DL, Zhang XJ, Shu YS, Zhang F, Jiang L, Song B.
evidence for supporting the mechanism of infrared and terahertz Demonstration of biophoton-driven DNA replication via gold
neurotransmission nanoparticle-distance modulated yield oscillation. Nano Res.
The assumption that consciousness can be understood as 2021;14(1):40–45.
a state of matter, quantum or photon, has led to some interesting 7. Tegmark M. Importance of quantum decoherence in brain
interdisciplinary questions, from neuroscience to materials processes. Phys Rev E. 2000;61(4):4194–4206.
science [5,6], condensed matter physics [17], and quantum 8. Liu X, Qiao Z, Chai Y, Zhu Z, Wu K, Ji W, Li D, Xiao Y, Mao L,
mechanics [18,19]. When the frequency of photon is equal to Chang C, et al. Nonthermal and reversible control of neuronal
the characteristic frequency of biological system, the photon signaling and behavior by midinfrared stimulation. Proc Natl
may nonthermally modulate molecular-related functions and Acad Sci USA. 2021;118(10):e2015685118.
biological processes by influencing the electronic states and 9. Zhang J, He Y, Liang S, Liao X, Li T, Qiao Z, Chang C, Jia H,
energies of biological macromolecules [3,12]. At the same Chen X. Non-invasive, opsin-free mid-infrared modulation
time, the rotational and vibrational energy levels of biological activates cortical neurons and accelerates associative learning.
macromolecules are mostly located in the terahertz band Nat Commun. 2021;12(1):2730.
[38–41]. In summary, our results confirm the possibility of 10. Mei WP. About the nature of biophotons. J Biol Syst.
using quantum theory to characterize biological processes and 1994;2(1):25–42.
the efficacy of THz photons for neural regulation, providing 11. Inaba H. Super-high sensitivity systems for detection and
a new pathway for intervention in neural signal generation spectral analysis of ultraweak photon emission from biological
and transmission. A deeper understanding of the mechanism cell cells and tissues. Experientia. 1988;44(7):550–559.
of biological information may provide us with subversive ideas 12. Salari V, Valian H, Bassereh H, Bókkon I, Barkhordari A.
and methods for developing new functional materials and Ultraweak photon emission in the brain. J Integr Neurosci.
exploring the unknown nature. 2015;14(4):419–429.
13. Thar R, Kühl M. Propagation of electromagnetic radiation in
mitochondria? J Theor Biol. 2004;230(2):261–270.
Acknowledgments 14. Zarkeshian P, Kumar S, Tuszynski J, Barclay P, Simon C. Are
there optical communication channels in the brain? Front
We thank X. Zhang of State Key Laboratory of Cognitive Biosci. 2018;23(8):1407–1421.
Neuroscience & Learning at Beijing Normal University for 15. Zhang F, Song B, Jiang L. The quantized chemical reaction
support during revision. C.C. acknowledges the support from resonantly driven by multiple MIR-photons: From nature to
the XPLORER Prize. Funding: This work was supported by the artificial. Nano Res. 2021;14(12):4367–4369.
the National Natural Science Foundation of China (No.12225511 16. Zhu Z, Chen C, Chang C, Song B. Terahertz-light induced
and T2241002), the National Defense Science and Technology structural transition and superpermeation of confined
Innovation Special Zone, and the National Supercomputer monolayer water. ACS Photonics 2021;8(3):781–786.

Tan et al. 2022 | https://doi.org/10.34133/research.0010 7


17. Zhang C, Yuan Y, Wu K, Wang Y, Zhu S, Shi J, Wang L, Li Q, 29. Guzman RE, Schwarz YN, Rettig J, Bruns D. SNARE
Zuo X, Fan C, et al. Driving DNA origami assembly with a force synchronizes synaptic vesicle fusion and controls
terahertz wave. Nano Lett. 2022;22(1):468–475. the kinetics of quantal synaptic transmission. J Neurosci.
18. Wu K, Qi C, Zhu Z, Wang C, Song B, Chang C. Terahertz wave 2010;30(31):10272–10281.
accelerates DNA unwinding: A molecular dynamics simulation 30. Zhang G, Huang S, Chaves A, Song C, Özçelik VO, Low T, Yan
study. J Phys Chem Lett. 2020;11(17):7002–7008. H. Infrared fingerprints of few-layer black phosphorus. Nat
19. Li Y, Chang C, Zhu Z, Sun L, Fan C. Terahertz wave enhances Commun. 2017;8:14071.
permeability of the voltage-gated calcium channel. J Am Chem 31. Schliesser A, Picqué N, Hänsch TW. Mid-infrared frequency
Soc. 2021;143(11):4311–4318. combs. Nat Photonics. 2012;6(7):440–449.
20. Liu G, Chang C, Qiao Z, Wu K, Zhu Z, Cui G, Peng W, Tang Y, 32. Camp CH Jr, Cicerone MT. Chemically sensitive bioimaging with
Li J, Fan C. Myelin sheath as a dielectric waveguide for signal coherent Raman scattering. Nat Photonics. 2015;9(5):295–305.
propagation in the mid-infrared to terahertz spectral range. 33. Gu M, Rondinelli JM. Linear and nonlinear optical probe of
Adv Funct Mater 2019;29(7):1807862. the ferroelectric-like phase transition in a polar metal, LiOsO3.
21. Xiang Z, Tang C, Chang C, Liu G. A new viewpoint and Appl Phys Lett. 2018;113(12):112903.
model of neural signal generation and transmission: 34. Kramer DM, Sacksteder CA. How acidic is the lumen?
Signal transmission on unmyelinated neurons. Nano Res. Photosynth Res. 1999;60(2):151–163.
2021;14(3):590–600. 35. Xi D, Xiao M, Cao J, Zhao L, Xu N, Long S, Fan J, Shao K,
22. Xiang ZX, Tang CX, Chang C, Liu GZ. A primary model Sun W, Yan X, et al. NIR light-driving barrier-free group
of THz and far-infrared signal generation and conduction rotation in nanoparticles with an 88.3% photothermal
in neuron systems based on the hypothesis of the ordered conversion efficiency for photothermal therapy. Adv Mater.
phase of water molecules on the neuron surface I: Signal 2020;32(11):1907855.
characteristics. Sci Bull. 2020;65(4):308–317. 36. Wu W, Yang Y, Yang Y, Yang Y, Zhang K, Guo L, Ge H,
23. Prince SM, Paulson AL, Jeong N, Zhang L, Amigues S, Chen X, Liu J, Feng H. Molecular engineering of an organic

Downloaded from https://spj.science.org on April 06, 2023


Singer AC. Alzheimer's pathology causes impaired inhibitory NIR-II fluorophore with aggregation-induced emission
connections and reactivation of spatial codes during spatial characteristics for in vivo imaging. Small. 2019;15(20):e1805549.
navigation. Cell Rep. 2021;35(3):109008. 37. Wang Z, Zhao J, Barbon A, Toffoletti A, Liu Y, An Y, Xu L,
24. Goda Y, Stevens CF. Two components of transmitter Karatay A, Yaglioglu HG, Yildiz EA, et al. Radical-enhanced
release at a central synapse. Proc Natl Acad Sci USA. intersystem crossing in new bodipy derivatives and application
1994;91(26):12942–12946. for efficient triplet-triplet annihilation upconversion. J Am
25. Watson JF, Pinggera A, Ho H, Greger IH. AMPA receptor Chem Soc. 2017;139(23):7831–7842.
anchoring at CA1 synapses is determined by N-terminal 38. Koala RASD, Fujita M, Nagatsuma T. Nanophotonics-inspired
domain and TARP γ8 interactions. Nat Commun. all-silicon waveguide platforms for terahertz integrated
2021;12(1):5083. systems. Nanophotonics 2022;11(9):1741–1759.
26. Dai J, Patzke C, Liakath-Ali K, Seigneur E, Südhof TC. GluD1 39. Globus T, Woolard D, Crowe TW, Khromova T, Gelmont
is a signal transduction device disguised as an ionotropic B, Hesler J. Terahertz Fourier transform characterization of
receptor. Nature. 2021;595(7866):261–265. biological materials in a liquid phase. J Phys D Appl Phys.
27. Gomez-Castro F, Zappettini S, Pressey JC, Silva CG, Russeau 2006;39(15):3405.
M, Gervasi N, Figueiredo M, Montmasson C, Renner M, 40. Li K, Li D, Zhang Y. Terahertz spectral properties of
Canas PM, et al. Convergence of adenosine and GABA 5-substituted uracils. Sensors, 2021;21(24):8292.
signaling for synapse stabilization during development. 41. Kowligy AS, Timmers H, Lind AJ, Elu U, Cruz FC,
Science. 2021;374(6568):eabk2055. Schunemann PG, Biegert J, Diddams SA. Infrared electric
28. Blum D, Lopes LV. Stabilizing synapses. Science. field sampled frequency comb spectroscopy. Sci Adv.
2021;374(6568):684–685. 2019;5(6):eaaw8794.

Tan et al. 2022 | https://doi.org/10.34133/research.0010 8

You might also like