You are on page 1of 8

Available online at www.sciencedirect.

com

St. Petersburg Polytechnical University Journal: Physics and Mathematics 1 (2015) 256–263
www.elsevier.com/locate/spjpm

The new method for studying neuronal activity: Optogenetics


Alexander I. Erofeev∗, Maxim V. Matveev, Stanislav G. Terekhin, Olga A. Zakharova,
Polina V. Plotnikova, Olga L. Vlasova
Peter The Great St. Petersburg Polytechnic University, 29 Politechnicheskaya St., St. Petersburg 195251, Russian Federation
Available online 4 December 2015

Abstract
The article is devoted to problems of realization and application of optogenetic methods used to identify reasons of various
diseases, to monitor the biochemical processes of cell activity and to study various organisms. The problems of delivery, embedding
and monitoring the expression of opsin genes into the cell genome of interest have been considered. In the article, the parameters and
properties of various opsins and also the main ways of achievement of precise optical control over cell using opsins were presented.
The rules for choosing the parameters of a light beam and the features of its putting were pointed out. The characteristic properties
of the different measurement technique and recording the experimental quantities were analyzed and given.

Copyright © 2015, St. Petersburg Polytechnic University. Production and hosting by Elsevier B.V.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Keywords: Optogenetics; Opsin; Photosensitivity; Channelrhodopsin; Halorhodopsin; Lentivirus; Transgenic mouse; Action potential; Fiber.

Introduction Importantly, this method entails creating genetically


coded constructions, i.e. optogenetic instruments which,
Optogenetics is a fundamentally new method of re- upon being delivered to specific cells, change their phys-
search that has been developed in the last decade in the iology when exposed to light. These instruments allow
Deisseroth Lab of Stanford University (Karl Deisseroth to control by light the electric activity of specific types of
is a professor of Bioengineering and of Psychiatry and neurons, cell signaling, and other processes. In order to
Behavioral Sciences). Optogenetics involves examining apply the method it is also necessary to create systems
how cells function by introducing photosensitive compo- for delivering light into tissues and for registering the
nents into their membranes; these components are capa- experimental results. While separate elements of these
ble of altering the properties of carrier cells in response to methods have existed since the 1970s, they were con-
being illuminated by a light beam of certain wavelength solidated to create optogenetic methods only in 2005
and thus act as the carrier’s fluorescent tags. [1]. Initially, the technology developed was aimed at
neuroscientific research. However, optogenetics turned
∗ Corresponding author. out to have a far wider potential. The method allows
E-mail addresses: alexandr.erofeew@gmail.com (A. I. to control certain events (with time resolutions on the
Erofeev), m.v.matveev@bk.ru (M. V. Matveev), stasok32@ order of milliseconds, which corresponds to the dura-
yandex.ru (S.G. Terekhin), ozakharpba92@gmail.com (O. tions of biological processes) in certain types of cells
A. Zakharova), plopolina@yandex.ru (P. V. Plotnikova),
olvlasova@yandex.ru (O. L. Vlasova).
[2–5].

http://dx.doi.org/10.1016/j.spjpm.2015.12.001
2405-7223/Copyright © 2015, St. Petersburg Polytechnic University. Production and hosting by Elsevier B.V. This is an open access article under
the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
(Peer review under responsibility of St. Petersburg Polytechnic University).
A. I. Erofeev et al. / St. Petersburg Polytechnical University Journal: Physics and Mathematics 1 (2015) 256–263 257

This analysis is particularly important since cellular of blue was derived from Volvox carteri algae [9]. By
events must be considered only in the context of other concurrently using several types of channelrhodopsins,
events occurring in individual tissues and in the body as an experimenter may simultaneously control mixed cell
a whole. populations: some commands may be given to cells of
the first type by yellow light, and others to the second
A brief history of the problem type by blue light. The so-called ‘fast’ and ‘slow’ opsins
were also created, allowing to control action potential du-
Francis Crick, the discoverer of the DNA structure, ration. The former opsins are capable of creating action
speculated in 1979 that one of the main problems in the potentials up to 200 times per second [10]. Opsins have
field of neurosciences is selectively controlling a spe- already been designed that are sensitive to light whose
cific type of brain cells given that the rest of the cells frequency is at the boundary between the visible and the
remain intact [6]. Since it is impossible to excite a spe- infra-red regions. Waves with this frequency penetrate
cific brain area with electrodes with the required preci- the tissues deeper and are more easily focused.
sion, and the effect of various medications is too slow, One of the most interesting possibilities of optoge-
Crick concluded that visible light has all the properties netic applications is controlling not only the electrical
enabling it to be used as an instrument of control. At that events in a neuron but also certain biochemical events.
time, however, there were no methods to make specific A lot of medicinal drugs are known to function through
cells photosensitive. the interaction with the family of membrane receptors
Even earlier, in 1971, Stoeckenius and Oesterhelt (GPCR). These receptors transduce external signals from
showed that bacteriorhodopsin acts as an ion pump some compound (medication) into cells, thus changing
that may be rapidly activated by visible-light photons the intracellular signaling, e.g., the calcium ion levels. If
[7]. Later other members of this family, halorhodopsin a photosensitive rhodopsin domain is added to a GPCR,
(1977) and channelrhodopsin (2002), were discovered it is possible to obtain receptors that are sensitive to green
[8]. light. These receptors have been termed optoXRs [11].
Still, the consensus for a long time had been that this When a single-component optoXR gene was delivered
combination of optical and genetic methods would not via a virus into the brains of laboratory animals, cell-
produce the desired effect: firstly, because the foreign specific control by light over certain biochemical signal
membrane proteins introduced to the cell could be toxic; transmission pathways was successfully exercised [11].
secondly, many scientists assumed the light-induced cur- Developing new fiber-optic instruments allowed to
rents to be too low. Additionally, to absorb photons, bac- deliver optical beams into any brain area of freely moving
terial opsins need a chemical cofactor, all-trans-retinal. animals. Additionally, a method allowing to simultane-
In the summer of 2005, a study was published demon- ously examine optical excitation and recording electrical
strating it was possible to use a bacterial opsin with- impulses was created. Currently it is possible, for exam-
out adding any other parts, components or reagents [1], ple, to directly measure the electrical activity in neural
with the neurons rendered photosensitive. In the follow- ensembles responsible for motor function while simul-
ing years, other researchers found that bacteriorhodopsin taneously controlling them through opsins.
and halorhodopsin, as well as channelrhodopsin, are ca- The first optogenetic studies on freely moving ani-
pable of turning neurons on and off rapidly and without mals were aimed at examining the neurons synthesiz-
any risks to cells in response to being irradiated by light ing the hypocretin neurotransmitter [12]. These cells are
of varying wavelengths. Vertebrate tissues already con- considered to be responsible for the narcolepsy sleep dis-
tain all-trans-retinal, and therefore optogenetic control is order. It was these cells that were discovered to exhibit
possible in intact brain tissues and even in freely moving specific types of electrical activity leading to awaken-
animals. ing. Optogenetics also helped prove that dopaminergic
neurons are responsible for the sense of joy [13].
Modern advances Research into the newest methods for treating Parkin-
son’s disease [14,15] has produced some of the best-
A number of supremely interesting experiments using known optogenetics experiments. This condition is char-
the new technology have been carried out in the recent acterized by impaired transmission of information in the
seven years. New opsins are under development with the Substantia nigra pars compacta neurons responsible for
goal of applying optogenetics to a wide array of stud- the motor function. Deep brain stimulation has been used
ies on various organisms. For example, in 2008, chan- to treat Parkinson’s disease since the 1990s. This proce-
nelrhodopsin VChR1 sensitive to yellow light instead dure involves alternating electrical pulses being sent to
258 A. I. Erofeev et al. / St. Petersburg Polytechnical University Journal: Physics and Mathematics 1 (2015) 256–263

specific brain areas using implanted tools. The potential six weeks may pass before a stable level of expression
efficiency of the treatment strategy is, however, severely in axonal distal ends is achieved.
limited, since the electrodes non-selectively stimulate
individual brain cells. A fundamental insight into this Electroporation
treatment method has been gained through optogenet-
ics. When different types of neurons were activated, un- A method of in utero electroporation can be used in
expected results were uncovered in parkinsonian mice. certain days of embryonic development. This method can
Apparently, the greatest therapeutic effort was achieved ensure targeted gene delivery to cortical layers I and III,
not by stimulating a certain type of cells but by control- and to striatal and hippocampal neurons [16–19]. Unlike
ling the activity of connecting axons. the viral methods, electroporation may deliver DNA of
any size with large promoter segments to achieve high
Genetic methods of opsin gene delivery to specific cellular specificity. Electroporation also allows to inject
neural populations numerous gene copies.

Opsin genes can be selectively expressed in a spe- Transgenic mice


cific pre-chosen type of neurons in the brain. Let us
discuss the main strategies whose efficiency has been The required opsin gene expression may be achieved
proved to achieve in vivo expression. One of the most by using transgene cassettes carrying recombinant pro-
widespread methods of genetic material delivery is using moters, and transgene constructs based on bacterial ar-
lentiviruses. Optimal gene expression time is two weeks tificial chromosomes. Several lines of transgenic mice
after injection. The main advantages of this method are expressing ChR2 driven by the Thy1 promoter were gen-
in a high level of gene expression and in the required ex- erated without altering their reproductive capacity or any
pression persisting for several years. The disadvantages discernible changes in behavior [20,21].
of using lentiviruses include insufficient specificity and a
low level of expression for several cell-specific promot- Cre-dependent expression systems
ers. Currently, gene material delivery by lentiviruses is
used in practically all experiments on mammals. Another Even though cell-specific promoters are effective for
common method is delivering the materials by adenoas- attaining the required expression levels in certain types of
sociated viruses (AAV). The optimal gene expression neurons, some promoters have weak transcriptional ac-
time is 3 weeks after injection. A less popular method tivity and it is thus impossible to use them to achieve the
is using Cre-dependent AAV expression systems. The level of expression where opsins may effectively induce
expression time is 3 weeks. The advantages and disad- the action potential. Special Cre-dependent AAV-vectors
vantages of the second and the third methods are similar were created to increase transcriptional activity. These
to those of the lentiviral method. vectors contain transgene cassettes expressing only in
the presence of Cre in transgenic animal lines. There-
Viral systems fore, in order to increase expression in a specific type of
cells, an appropriate line of mice must be chosen.
In contrast with most of the genetic methods,
lentivirus- and AAV-based viral vectors do not require Main strategies of optical control by opsins
using transgenic animal models. These methods allow
obtaining a high opsin gene expression level in both ro- Let us discuss the basic experimental methods for
dents and primates for periods of up to several months. achieving optical control using opsins.
Lentivirus- and AAV-based vectors used in experiments
have the following parameters: Rapid excitation – channelrhodopsins(ChR)

1. there are over 109 transducing units for lentiviruses; In some cases the bacterial rhodopsin gene introduced
2. there are over 1012 genome copies for AAV. into neurons may cause a light-induced photocurrent.
Currently, modified opsins are mainly used. In particu-
Generally, opsin gene expression in the rodent brain lar, some codons of algae were replaced by codons of
reaches the required level in three weeks after AAV in- mammals, which significantly increased the expression
jection and in two weeks after lentivirus injection. Over of these genes. Of course, changing codons may cause
A. I. Erofeev et al. / St. Petersburg Polytechnical University Journal: Physics and Mathematics 1 (2015) 256–263 259

unforeseen effects, when along with an increased expres- successfully proved in a model of freely moving worms,
sion in mammalian neurons, there is a depression of some in thin sections from the mammalian brain [35], and also
other function or a decrease in the expression in other in a neuronal culture [36], it took a few years to achieve
cell types. For example, introducing the H134R mutant similar results for intact mammals due to problems with
into ChR2 lead to a twofold increase in photocurrent dur- membrane transport necessitating the use of additional
ing prolonged stimulation, while the temporal precision methods.
sharply reduced as the channel closing rate decreased Opsin eNpHR2.0 was engineered by various meth-
[22]. ods; it is characterized by higher current values [37,21].
A significant red shift in the optical absorption spec- This allowed to use it for intact rodent tissues [38], as
trum of algal protein VChR1 [23] stimulated with yel- well as for primate and human tissues [39]. Ultimately,
low light (excitation wavelength of 590 nm) without eNpHR3.0 was created with even higher photocurrent
having any effect on ChR2 (excitation wavelength of values for a moderate light intensity in the yellow range
470 nm) permits combined excitation control in vivo. or red-shifted (up to 680 nm) [26].
Most channelrhodopsins studied to date have a rela- It should be noted that it is better to use bidirectional
tively low single-channel conductance and broad cation control for experiments in neuronal function inhibition
selectivity. Combined methods, including those using and to take into account the stability of the photocur-
VCHR1 , allow obtaining various characteristics of cel- rents of inhibitory opsins. Finally, doses of light expo-
lular photocurrents. sure should be carefully managed, in particular, in order
For example, a study of L132Cmutant [24] revealed to prevent tissue overheating during constant exposure.
that its activation leads to insignificant photocurrents This is why it is important to control the light intensity
of calcium ions at physiological concentrations of cal- required for inhibition [40].
cium, and the increase in cytosolic Ca2+ concentration
is mainly engendered by the activation of endogenous Step-function opsins (SFO)
voltage-gated calcium channels due to neuronal mem-
brane depolarization [25]. Second- and even third-order Conductance of wild-type ChR2 deactivates after
conductance levels should always be taken into account, photostimulation stops (it lasts about 10 ms), while
especially when a high conductance of calcium ions is mutant ChR2 (C128X) (mutations to cysteine-128 and
observed. aspartate-156) deactivates far more slowly [27,28]. For
Different types of cells, and even different cellular example, C128T, C128A and C128S-substituted mutant
areas may induce, transport, or respond to increased cal- proteins are characterized by reaction times of 2, 42 and
cium concentrations in different ways. The latest mod- 100 s, respectively [27]. This stable blue light-induced
eling studies where cellular responses to photostimula- photocurrent can be stopped by yellow light pulses (560–
tion were integrated with the Hodgkin–Huxley model 590 nm). Mutant genes of this class are called step-
[26] may be expanded and complemented with second- function opsins (SFO), since they allow step control the
order conductances, which means that it is now possible membrane potential. This type of control will likelier
to predict cellular responses to various photostimulation lead to an action potential threshold and increase the
methods. probability of endogenous synaptic output [27].

Rapid inhibition – halorhodopsins (NpHR) Key differences between SFO and ChR

Inhibition also plays an important role in optogenetic Let us mention two of these differences. Firstly, the
studies of the activity of neurons and neural ensembles. cellular photosensitivity of SFO is higher than that of
Halorhodopsin (HR) inducing electrogenic Cl− current ChR; this is a result of open channel accumulation dur-
is used for these studies, but it typically has a desensitiz- ing a light pulse [27,29]. The second difference between
ing effect. However, a Natronomonas pharaonis homo- SFO and ChR is driven by an asynchronous nature of
logue gene [32–34] is capable of inducing a stable pho- SFO-mediated neuronal activity that does not engage all
tocurrent [35] with a wavelength maximum at 590 nm expressing neurons into a united pattern caused by pho-
(excitation at this wavelength does not elicit any response tostimulation.
from ChR2 , which allows to activate ChR2 and NpHR There currently exist SFOs with deactivation times
independent of bidirectional activity modulation). Un- up to 30 min [30]; this allows to bring the expressing
like excitatory ChR, NpHR requires constant light ex- neurons to a stable resting potential with subsequent
posure. Even though NpHR-assisted inhibition was removal of the light source, i.e. allows to conduct the
260 A. I. Erofeev et al. / St. Petersburg Polytechnical University Journal: Physics and Mathematics 1 (2015) 256–263

subsequent behavioral or physiological experiment with- neurons and neuronal ensembles function, experimen-
out any light or other equipment. Moreover, using pro- tal results, generally, are strongly dependent on types
longed low-intensity light pulses removes the hetero- of neurons and stimulation parameters (frequency, dura-
geneities in the response of the expressing cells. In this tion, amplitude, etc.). Choice of opsin (e.g., H134R or
case, even large volumes of tissues may eventually be L132C) is also of substantial importance.
driven to saturation levels. A huge variety of genes of microbial opsins are found
However, despite the wide experimental potential of in nature, which offers ample opportunities for designing
SFOs, their use must always be accompanied by addi- new optogenetic tools.
tional electrophysiological studies. This is necessary to
correctly interpret the obtained data. Tools for regulating biochemical signaling
But even though the above-listed ChRs have many
useful properties, none of them are capable of produc- The above-described microbial opsin genes (type I)
ing spike trains with frequencies above 40 Hz, while encode ion channels that control the excitability of neu-
many types of neurons and their physiological processes rons by modifying their membrane potential above or
require high-frequency spike trains (over 40 Hz). Even below the action potential generation threshold. While
with wild-type ChR2 (10 ms), and also H134R (20 ms), the advantages of this approach are in its speed and pre-
precision control is limited at high frequencies. Substi- cision, some experiments require temporal and precise
tuting the Glu-123 residue with threonine or alanine lead modulation of intracellular processes.
to the acceleration of the channel closing kinetics from There is one more type of opsins (type II), for ex-
10 to 4 ms due to a moderate decrease in photocurrent, ample, the photosensitive proteins in mammalian eyes;
which significantly improved the precision of optoge- these proteins are capable of not only inducing a pho-
netic control [10]. The E123 mutants are unique among tocurrent at light exposure but of acting as G-coupled
ChRs as they eliminate the sensitivity of channel ki- protein receptors (GPCRs), and therefore taking part in
netics to membrane potentials, no matter whether they intracellular signaling. It is possible to control slow in-
act separately or in combination with other H134R and hibition [41] or excitation [42]. Currently a great num-
T159C mutants [10,31]. After these non-linear and non- ber of chimeras [43] between vertebrate rhodopsins and
stationary effects are removed, it becomes easier to pre- GPCR families are being developed that may serve as
dict and model the channel response. Opsins of this class one-component control tools (among them, dopaminer-
(separate E123 mutants or combinations of other mutants gic, serotonergic and adrenergic receptors which all play
[10]) are called ChETAs (ChRE123T/A). ChETAs can an important role in neurotransmission and neuromodu-
be used not only for neurons requiring high-frequency lation). These optogenetic tools are called optoXRs, and
spike trains, since they help reduce the amount of extra they allow to control intracellular signaling for studying
spikes along with the falsely prolonged depolarization. the behavior of freely moving mice [11].
ChETAs have been shown to provide enhanced perfor- The speed and precision achieved through methods
mance within intact tissues of mammalian brains [10]; at of biochemical optogenetics provide opportunities that
the same time, fast deactivation typically leads to a de- are unattainable by pharmacological or genetic methods.
crease in effective cellular sensitivity to prolonged light The active development of this area of optogenetics al-
pulses, as less channels remain open. lows to use these technologies for practically all types of
cells.
Pharmacological, optogenetic and electric stimulation
Selecting light beam parameters and light delivery
These types of stimulation differ from the natural
synaptic transmission on account of the changes in the After opsin expression was achieved in neurons pre-
conductance of ion flows and membrane potentials. Any senting an interest to researchers, the problem of light
of these stimulation types may affect intracellular mem- beam delivery arose. The requirements imposed on the
branes, endoplasmic reticula, nuclear complexes, synap- beam’s parameters vary depending on the conditions of
tic vesicles and mitochondria. These factors need to be the experiment. For example, beam parameters required
taken into account, especially when studying single neu- for studying fast oscillations in thin brain slices while
rons. Despite the novelty, the precision and the specificity using several opsins in vitro are different from those for
of optogenetic methods, their results must be compared studying the effects of prolonged in vivo stimulation of
to electric stimulation results under similar conditions. certain regions of the animal brain [44]. The parame-
Even though optogenetics allows to explain exactly how ters of photocurrents induced in neurons by light pulses
A. I. Erofeev et al. / St. Petersburg Polytechnical University Journal: Physics and Mathematics 1 (2015) 256–263 261

depend on many factors. Some of them are the type of of photons per surface unit, per time unit. However, an-
expressed opsin, irradiation wavelength, intensity and other quantity is more convenient to use in experiments;
duration, and even events that happened before the start it is the luminous flux density measured in mW/mm2
of irradiation. If not all channelrhodopsin molecules re- and defined as the luminous flux multiplied by a pho-
cover their initial state after the previous exposure, the ton energy. For a wild-type channelrhodopsin ChR2 with
initial response to a light pulse is reduced. standard levels of expression and light illumination at a
473-nm wavelength, the flux density necessary for initi-
Activation parameters for various opsins ating the axonal potential is 1–5 mW/mm2 .
As previously noted, the requirements for the duration
For the ChR2 , ChR2 (H134R), ChR1/2 chimeras, of illumination depend on the conditions of various op-
ChETA (derived from Chlamydomonas reinhardtii) togenetic experiments. In case of optogenetic inhibition
opsin family, the excitation wavelength is 470 nm, while the time range is determined by the duration of this inhi-
the main function of these opsins is cellular membrane bition (flux density of 1–5 mW/mm2 ), while for bistable
depolarization. This opsin family is capable of fast on/off optogenetic control a short period of time is required,
switching, which is why these opsins are best suited for and the light beam must have a far lower flux density
precise activation of neurons. Substituting H134R results (less than 0.01 mW/mm2 ).
in more photocurrents compared to wild-type ChR2 s. In For in vitro experiments, when a tissue sample is ob-
particular, the ChR1 /ChR2 chimeras and ChETA produce served under a microscope, the most appropriate light
spikes up to 200 Hz. sources, i.e., halogen/xenon lamps, LEDs, lasers, can be
The next group of opsins are the step-functional placed directly along the microscope beam. For some ex-
opsins (SFOs): ChR2 (C128A), ChR2 (C128S), ChR2 periments pulsed illumination is required. Fast shutters
(C128T) (derived from Chlamydomonas reinhardtii). (e.g., Lambda DG-4 or Uniblitzshutter) or pulse lasers
The excitation wavelength is 470 nm, and a light beam can be used to generate short light pulses. High-power
with a 546-nm wavelength is used for inhibition. The sources (10–15 mW at the tip of a 100 μm fiber) are
main function of these opsins is also cellular membrane best suited for in vivo studies on freely moving ani-
depolarization. ChR2 s (with point mutations) are char- mals. LEDs can also be used to stimulate cortical layers,
acterized by slow or optically switchable deactivation. while thin optical fibers must be used to control deeper
Substituting C128A and C128S result in the longest ac- brain regions. Optical fibers are used for fabricating the
tivation and the highest photosensitivity, while C128T so-called optrodes which are instruments for simulta-
retains a high temporal precision. SFOs can be switched neously recording electrophysiological parameters and
on and off by blue and green light pulses. optically exciting opsins. Fiber thickness is also cho-
VChR1 (derived from Volvox carteri) has an excita- sen depending on the nature of the studied object. For
tion wavelength of 535 nm, and its main function is also example, for mice with unrestricted movement, the re-
cellular membrane depolarization. A red shift in the ac- quired thickness is no more than 300 μm, and for rats it is
tivation spectrum (relative to ChR2 ) allows to exercise 400 μm. The fact that mammalian brain tissues strongly
combined control. absorb light should also be kept in mind when setting
NpHR, eNpHR (derived from Natronomonas pharao- up experiments; for example, light beam intensity at a
nis) have an excitation wavelength of 589 nm, but their 500 μm distance from the fiber tip is about 10% of the
main function is cellular membrane hyperpolarization. initial one.
This group of opsins is, in a manner of speaking, a light- Currently the Laboratory of Molecular Neurodegen-
activated pump for chloride ions. These opsins are used eration is developing a hardware and software pack-
to hyperpolarize neurons with high temporal precision age for optogenetic research. The proposed package is
and stable inhibition over several minutes. a source of light emission consisting of several bright
Opto-α1AR, opto-β2AR (obtained synthetically) LEDs, with special software with the required ex-
have an excitation wavelength of 500 nm; their main perimentally chosen diode parameters for opsins pro-
function is biochemical control over the cellular mem- grammed in.
brane. This group of opsins includes light-activated
GPCRs (activated via the G-protein). Methods of recording the experimental data
When designing a system of light delivery for opsin
activation, the absorption coefficient for photons with a Various methods of measuring experimental param-
certain wavelength must be taken into account. It is pro- eters are used for optogenetic control. First of all, these
portional to the luminous flux which in turn is the number are the methods of obtaining and analyzing images
262 A. I. Erofeev et al. / St. Petersburg Polytechnical University Journal: Physics and Mathematics 1 (2015) 256–263

using various dyes which include Ca2+ -specific dyes The part of the study dedicated to staging and testing
(e.g., fura-3, Fluo-5F), and also voltage-sensitive dyes optogenetic methods is supported by the Russian Scien-
(VSDs, e.g., RH-155). Such methods are effective for tific Fund grant no. 14-25-0024.
measuring the electrical activity in large cell popula-
tions ex vivo and in vivo with a high temporal res-
olution. A two-photon microscope can be used for References
imaging with Ca2+ -specific dyes, since noises from
channelrhodopsin photoactivation are virtually absent [1] E.S. Boyden, F. Zhang, E. Bamberg, et al., Millisecond-timescale,
genetically targeted optical control of neural activity, Nat. Neu-
during two-photon excitation. Voltage-sensitive dyes are rosci. 8 (9) (2005) 1263–1268.
lipophilic molecules whose optical absorption depends [2] K. Deisseroth, G. Feng, A.K. Majewska, et al., Next-generation
on membrane potential. Along with high-speed cameras optical technologies for illuminating genetically targeted brain
for recording the changes in optical signal, VSD imaging circuits, J. Neurosci. 26 (41) (2006) 10380–10386.
is used, which allows to detect the changes in neural elec- [3] M. Scanziani, M. Häusser, Electrophysiology in the age of light,
Nature 461 (7266) (2009) 930–939.
trical activity with high spatial and temporal resolutions [4] K. Deisseroth, Controlling the brain with light, Sci. Am 303 (5)
(on the order of μm and ms). The absorption maximum (2010) 48–55.
for the RH-155 dye is 700 μm, while excitation peaks [5] K. Deisseroth, Optogenetics, Nat. Methods 8 (1) (2011) 26–29.
for opsins are in the 470–590 μm range. Such a differ- [6] F.H. Crick, Thinking about the brain, Sci. Am. 241 (3) (1979)
ence in wavelengths allows to simultaneously optically 219–232.
[7] D. Oesterhelt, W. Stoeckenius, Rhodopsin-like protein from the
stimulate opsins and to detect images. purple membrane of Halobacterium halobium, Nat. New Biol.
Another class of methods for measuring the pa- 233 (39) (1971) 149–152.
rameters of optogenetic experiments entails simultane- [8] G. Nagel, D. Ollig, M. Fuhrmann, et al., Channelrhodopsin-1:
ous control of animal behavior and electrophysiological a light-gated proton channel in green algae, Science 296 (5577)
recording. For this goal, special optical fiber-based in- (2002) 2395–2398.
[9] F. Zhang, M. Prigge, F. Beyriere, et al., Red-shifted optogenetic
struments were designed to deliver light beams into the excitation: a tool for fast neural control derived from Volvox car-
expression area of opsin genes and for electrophysiolog- teri, Nat. Neurosci. 11 (6) (2008) 631–633.
ical measurements. [10] L.A. Gunaydin, O. Yizhar, A. Berndt, et al., Ultrafast optogenetic
control, Nat. Neurosci. 13 (3) (2010) 387–392.
[11] R.D. Airan, K.R. Thompson, L.E. Fenno, et al., Temporally pre-
cise in vivo control of intracellular signaling, Nature 458 (7241)
Conclusion
(2009) 1025–1029.
[12] A.R. Adamantidis, F. Zhang, A.M. Aravanis, et al., Neural sub-
In this article we detailed the gist of the optogenetic strates of awakening probed with optogenetic control of hypocre-
method, its main components and application areas. This tin neurons, Nature 450 (7168) (2007) 420–424.
method is currently being rapidly developed and im- [13] H.C. Tsai, F. Zhang, A. Adamantidis, et al., Phasic firing in
dopaminergic neurons is sufficient for behavioral conditioning,
proved, and applied in an increasing number of scientific
Science 324 (5930) (2009) 1080–1084.
areas. Taking the optogenetic approach to studying vari- [14] V. Gradinaru, M. Mogri, K.R. Thompson, et al., Optical decon-
ous neurodegenerative diseases is of the greatest interest struction of parkinsonian neural circuitry, Science 324 (5925)
to the authors. For example, in 2013, the abnormalities of (2009) 354–359.
synaptic transmission in a cortical and striatal neuronal [15] A.V. Kravitz, B.S. Freeze, P.R. Parker, et al., Regulation of parkin-
sonian motor behaviours by optogenetic control of basal ganglia
culture were demonstrated on mouse models of Hunting-
circuitry, Nature 466 (7306) (2010) 622–626.
ton’s disease using the optogenetic method [45]. Study- [16] H. Adesnik, M. Scanziani, Lateral competition for cortical space
ing this type of problem is the subject of the authors’ by layer-specific horizontal circuits, Nature 464 (7292) (2010)
current and future research. 1155–1160.
[17] V. Gradinaru, K.R. Thompson, F. Zhang, et al., Targeting and
readout strategies for fast optical neural control in vitro and in
vivo, J. Neurosci. 27 (52) (2007) 14231–1438.
Acknowledgment [18] T.L. Lewis, T. Mao, K. Svoboda, D.B. Arnold, Myosin-dependent
targeting of transmembrane proteins to neuronal dendrites, Nat.
The authors express their gratitude to I.B. Bezproz- Neurosci. 12 (5) (2009) 568–576.
vanny, the head of the Laboratory of Molecular Neu- [19] L. Petreanu, D. Huber, A. Sobczyk, K. Svoboda,
Channelrhodopsin-2-assisted circuit mapping of long-range
rodegeneration, doctor of biological sciences and Carl
callosal projections, Nat. Neurosci. 10 (5) (2007) 663–668.
J. and Hortense M. Thomsen Chair in Alzheimer’s Dis- [20] B.R. Arenkiel, J. Peca, I.G. Davison, et al., In vivo light-
ease Research, and to the whole team of the laboratory induced activation of neural circuitry in transgenic mice express-
for consultations and assistance in research. ing channelrhodopsin-2, Neuron 54 (2) (2007) 205–218.
A. I. Erofeev et al. / St. Petersburg Polytechnical University Journal: Physics and Mathematics 1 (2015) 256–263 263

[21] S. Zhao, C. Cunha, F. Zhang, Q. Liu, B. Gloss, Improved ex- [35] F. Zhang, L.P. Wang, M. Brauner, et al., Multimodal fast optical
pression of halorhodopsin for light-induced silencing of neuronal interrogation of neural circuitry, Nature 446 (7136) (2007) 633–
activity, Brain Cell Biol. 36 (1-4) (2008) 141–154. 639.
[22] G. Nagel, M. Brauner, J.F. Liewald, et al., Light activation of [36] X. Han, E.S. Boyden, Multiple-color optical activation, silenc-
channelrhodopsin-2 in excitable cells of Caenorhabditis elegans ing, and desynchronization of neural activity, with single-spike
triggers rapid behavioral responses, Curr. Biol. 15 (24) (2005) temporal resolution, Plos One 2 (3) (2007) e299.
2279–2284. [37] V. Gradinaru, K.R. Thompson, K. Deisseroth, eNpHR: a Na-
[23] F. Zhang, M. Prigge, F. Beyriere, et al., Red-shifted optogenetic tronomonas halorhodopsin enhanced for optogenetic applica-
excitation: a tool for fast neural control derived from Volvox car- tions, Brain Cell Biol. 36 (1-4) (2008) 129–139.
teri, Nat. Neurosci. 11 (6) (2008) 631–633. [38] V.S. Sohal, F. Zhang, O. Yizhar, K. Deisseroth, Parvalbumin neu-
[24] S. Kleinlogel, K. Feldbauer, R.E. Dempski, et al., Ultra light- rons and gamma rhythms enhance cortical circuit performance,
sensitive and fast neuronal activation with the Ca2+-permeable Nature 459 (7247) (2009) 698–702.
channelrhodopsin CatCh, Nat. Neurosci. 14 (4) (2011) 513–518. [39] V. Busskamp, J. Duebel, D. Balya, et al., Genetic reactivation of
[25] Y.P. Zhang, T.G. Oertner, Optical induction of synaptic plasticity cone photoreceptors restores visual responses in retinitis pigmen-
using a light-sensitive channel, Nat. Methods 4 (2) (2007) 139– tosa, Science 329 (5990) (2010) 413–417.
141. [40] A.M. Aravanis, L.P. Wang, F. Zhang, et al., An optical neural
[26] V. Gradinaru, F. Zhang, C. Ramakrishnan, et al., Molecular and interface: in vivo control of rodent motor cortex with integrated
cellular approaches for diversifying and extending optogenetics, fiberoptic and optogenetic technology, J. Neural Eng. 4 (3) (2007)
Cell 141 (1) (2010) 154–165. 143–156.
[27] A. Berndt, O. Yizhar, L.A. Gunaydin, et al., Bi-stable neural state [41] X. Li, D.V. Gutierrez, M.G. Hanson, et al., Fast noninvasive acti-
switches, Nat. Neurosci. 12 (2) (2009) 229–234. vation and inhibition of neural and network activity by vertebrate
[28] C. Bamann, R. Gueta, S. Kleinlogel, et al., Structural guidance of rhodopsin and green algae channelrhodopsin, Proc. Natl. Acad.
the photocycle of channelrhodopsin-2 by an interhelical hydrogen Sci. 102 (49) (2005) 17816–17821.
bond, Biochemistry 49 (2) (2010) 267–278. [42] Z. Melyan, E.E. Tarttelin, J. Bellingham, et al., Addition of human
[29] I. Diester, M.T. Kaufman, M. Mogri, et al., An optogenetic tool- melanopsin renders mammalian cells photoresponsive, Nature
box designed for primates, Nat. Neurosci. 14 (3) (2011) 387–397. 433 (7027) (2005) 741–745.
[30] O. Yizhar, L. Fenno, M. Prigge, et al., Neocortical excita- [43] J.M. Kim, J. Hwa, P. Garriga, et al., Light-driven activation of beta
tion/inhibition balance in social dysfunction and information pro- 2-adrenergic receptor signaling by a chimeric rhodopsin contain-
cessing, Nature 477 (7363) (2011) 171–178. ing the beta 2-adrenergic receptor cytoplasmic loops, Biochem-
[31] A. Berndt, P. Schoenenberger, J. Mattis, et al., High-efficiency istry 44 (7) (2005) 2284–2292.
channelrhodopsins for fast neuronal stimulation at low light lev- [44] T. Bruegmann, D. Malan, M. Hesse, et al., Optogenetic control
els, Proc. Natl. Acad. Sci. 108 (18) (2011) 7595–7600. of heart muscle in vitro and in vivo, Nat. Methods 7 (11) (2010)
[32] J.K. Lanyi, D. Oesterhelt, Identification of the retinal-binding 897–900.
protein in halorhodopsin, J. Biol. Chem. 257 (5) (1982) 2674– [45] D.N. Artamonov, V.V. Korzhova, D. Vu, et al., Violations of
2677. synaptic transmission in Huntington’s disease in the cortico-
[33] B. Scharf, M. Engelhard, Blue halorhodopsin from Natronobac- striatal neuronal culture models, Biochem. (Mosc.) 30 (4) (2013)
terium pharaonis: wavelength regulation by anions, Biochemistry 1–13.
33 (21) (1994) 6387–6393.
[34] M. Sato, M. Kubo, T. Aizawa, et al., Role of putative anion-
binding sites in cytoplasmic and extracellular channels of
Natronomonas pharaonis halorhodopsin, Biochemistry 44 (12)
(2005) 4775–4784.

You might also like