Vous êtes sur la page 1sur 116

Optogenetics

Optogenetics (from Greek optikós, meaning


'seen, visible') is a biological technique which
involves the use of light to control cells in living
tissue, typically neurons, that have been
genetically modified to express light-sensitive
ion channels. It is a neuromodulation method
that uses a combination of techniques from
optics and genetics to control and monitor the
activities of individual neurons in living tissue—
even within freely-moving animals—and to
precisely measure these manipulation effects in
real-time.[1] The key reagents used in
optogenetics are light-sensitive proteins.
Neuronal control is achieved using optogenetic
actuators like channelrhodopsin, halorhodopsin,
and archaerhodopsin, while optical recording of
neuronal activities can be made with the help of
optogenetic sensors for calcium (GCaMP),
vesicular release (synapto-pHluorin),
Neurotransmitter (GluSnFRs), or membrane
voltage (Arc Lightning, ASAP1).[2] [3] Control (or
recording) of activity is restricted to genetically
defined neurons and performed in a
spatiotemporal-specific manner by light.

In 2010, optogenetics was chosen as the


"Method of the Year" across all fields of science
and engineering by the interdisciplinary
research journal Nature Methods.[4] At the same
time, optogenetics was highlighted in the article
on "Breakthroughs of the Decade" in the
academic research journal Science.[5] These
journals also referenced recent public-access
general-interest video Method of the year video
and textual SciAm summaries of optogenetics.

History
The "far-fetched" possibility of using light for
selectively controlling precise neural activity
(action potential) patterns within subtypes of
cells in the brain was thought of by Francis
Crick in his Kuffler Lectures at the University of
California in San Diego in 1999.[6] An earlier use
of light to activate neurons was carried out by
Richard Fork,[7] who demonstrated laser
activation of neurons within intact tissue,
although not in a genetically-targeted manner.
The earliest genetically targeted method that
used light to control rhodopsin-sensitized
neurons was reported in January 2002, by Boris
Zemelman (now at UT Austin) and Gero
Miesenböck, who employed Drosophila
rhodopsin cultured mammalian neurons.[8] In
2003, Zemelman and Miesenböck developed a
second method for light-dependent activation
of neurons in which single inotropic channels
TRPV1, TRPM8 and P2X2 were gated by
photocaged ligands in response to light.[9]
Beginning in 2004, the Kramer and Isacoff
groups developed organic photoswitches or
"reversibly caged" compounds in collaboration
with the Trauner group that could interact with
genetically introduced ion channels.[10][11]
TRPV1 methodology, albeit without the
illumination trigger, was subsequently used by
several laboratories to alter feeding, locomotion
and behavioral resilience in laboratory
animals.[12][13] [14] However, light-based
approaches for altering neuronal activity were
not applied outside the original laboratories,
likely because the easier to employ
channelrhodopsin was cloned soon
thereafter.[15]

Peter Hegemann, studying the light response of


green algae at the University of Regensbug, had
discovered photocurrents that were too fast to
be explained by the classic g-protein-coupled
animal rhodopsins.[16] Teaming up with the
electrophysiologist Georg Nagel at the Max
Planck Institute in Frankfurt, they could
demonstrate that a single gene from the alga
Chlamydomonas produced large photocurents
when expressed in the oocyte of a frog.[17] To
identify expressing cells, they replaced the
cytoplasmic tail of the algal protein with the
fluorescent protein YFP, generating the first
generally applicable optogenetic tool.[15] Zhuo-
Hua Pan of Wayne State University, researching
on restore sight to blindness, thought about
using channelrhodopsin when it came out in
late 2003. By February 2004, he was trying
channelrhodopsin out in ganglion cells — the
neurons in our eyes that connect directly to the
brain — that he had cultured in a dish. Indeed,
the transfected neurons became electrically
active in response to light.[18] In April 2005,
Susana Lima and Miesenböck reported the first
use of genetically-targeted P2X2
photostimulation to control the behaviour of an
animal.[19] They showed that photostimulation
of genetically circumscribed groups of neurons,
such as those of the dopaminergic system,
elicited characteristic behavioural changes in
fruit flies. In August 2005, Karl Deisseroth's
laboratory in the Bioengineering Department at
Stanford including graduate students Ed
Boyden and Feng Zhang (both now at MIT)
published the first demonstration of a single-
component optogenetic system in cultured
mammalian neurons,[20][21] using the
channelrhodopsin-2(H134R)-eYFP construct
from Nagel and Hegemann.[15] The groups of
Gottschalk and Nagel were first to use
Channelrhodopsin-2 for controlling neuronal
activity in an intact animal, showing that motor
patterns in the roundworm Caenorhabditis
elegans could be evoked by light stimulation of
genetically selected neural circuits (published in
December 2005).[22] In mice, controlled
expression of optogenetic tools is often
achieved with cell-type-specific Cre/loxP
methods developed for neuroscience by Joe Z.
Tsien back in 1990s[23] to activate or inhibit
specific brain regions and cell-types in vivo.[24]

The primary tools for optogenetic recordings


have been genetically encoded calcium
indicators (GECIs). The first GECI to be used to
image activity in an animal was cameleon,
designed by Atsushi Miyawaki, Roger Tsien and
coworkers.[25] Cameleon was first used
successfully in an animal by Rex Kerr, William
Schafer and coworkers to record from neurons
and muscle cells of the nematode C.
elegans.[26] Cameleon was subsequently used
to record neural activity in flies[27] and
zebrafish.[28] In mammals, the first GECI to be
used in vivo was GCaMP,[29] first developed by
Nakai and coworkers.[30] GCaMP has
undergone numerous improvements, and
GCaMP6[31] in particular has become widely
used throughout neuroscience.

In 2010, Karl Deisseroth at Stanford University


was awarded the inaugural HFSP Nakasone
Award "for his pioneering work on the
development of optogenetic methods for
studying the function of neuronal networks
underlying behavior". In 2012, Gero Miesenböck
was awarded the InBev-Baillet Latour
International Health Prize for "pioneering
optogenetic approaches to manipulate neuronal
activity and to control animal behaviour." In
2013, Ernst Bamberg, Ed Boyden, Karl
Deisseroth, Peter Hegemann, Gero Miesenböck
and Georg Nagel were awarded The Brain Prize
for "their invention and refinement of
optogenetics."[32][33] Karl Deisseroth was
awarded the Else Kröner Fresenius Research
Prize 2017 (4 million euro) for his "contributions
to the understanding of the biological basis of
psychiatric disorders".

Description
Fig 1. Channelrhodopsin-2 (ChR2) induces temporally precise
blue light-driven activity in rat prelimbic prefrontal cortical
neurons. a) In vitro schematic (left) showing blue light
delivery and whole-cell patch-clamp recording of light-evoked
activity from a fluorescent CaMKllα::ChR2-EYFP expressing
pyramidal neuron (right) in an acute brain slice. b) In vivo
schematic (left) showing blue light (473 nm) delivery and
single-unit recording. (bottom left) Coronal brain slice
showing expression of CaMKllα::ChR2-EYFP in the prelimbic
region. Light blue arrow shows tip of the optical fiber; black
arrow shows tip of the recording electrode (left). White bar,
100 µm. (bottom right) In vivo light recording of prefrontal
cortical neuron in a transduced CaMKllα::ChR2-EYFP rat
showing light-evoked spiking to 20 Hz delivery of blue light
pulses (right). Inset, representative light-evoked single-unit
response.[34]
Fig 2. Halorhodopsin (NpHR) rapidly and reversibly silences
spontaneous activity in vivo in rat prelimbic prefrontal cortex.
(Top left) Schematic showing in vivo green (532 nm) light
delivery and single- unit recording of a spontaneously active
CaMKllα::eNpHR3.0- EYFP expressing pyramidal neuron.
(Right) Example trace showing that continuous 532 nm
illumination inhibits single-unit activity in vivo. Inset,
representative single unit event; Green bar, 10 seconds.[34]
Play media
A nematode expressing the light-sensitive ion channel Mac.
Mac is a proton pump originally isolated in the fungus
Leptosphaeria maculans and now expressed in the muscle
cells of C. elegans that opens in response to green light and
causes hyperpolarizing inhibition. Of note is the extension in
body length that the worm undergoes each time it is exposed
to green light, which is presumably caused by Mac's muscle-
relaxant effects.[35]

Play media
A nematode expressing ChR2 in its gubernacular-oblique
muscle group responding to stimulation by blue light. Blue
light stimulation causes the gubernacular-oblique muscles to
repeatedly contract, causing repetitive thrusts of the spicule,
as would be seen naturally during copulation.[36]
Optogenetics provides millisecond-scale
temporal precision which allows the
experimenter to keep pace with fast biological
information processing (for example, in probing
the causal role of specific action potential
patterns in defined neurons). Indeed, to probe
the neural code, optogenetics by definition must
operate on the millisecond timescale to allow
addition or deletion of precise activity patterns
within specific cells in the brains of intact
animals, including mammals (see Figure 1). By
comparison, the temporal precision of
traditional genetic manipulations (employed to
probe the causal role of specific genes within
cells, via "loss-of-function" or "gain of function"
changes in these genes) is rather slow, from
hours or days to months. It is important to also
have fast readouts in optogenetics that can
keep pace with the optical control. This can be
done with electrical recordings ("optrodes") or
with reporter proteins that are biosensors,
where scientists have fused fluorescent
proteins to detector proteins. An example of
this is voltage-sensitive fluorescent protein
(VSFP2).[37] Additionally, beyond its scientific
impact optogenetics represents an important
case study in the value of both ecological
conservation (as many of the key tools of
optogenetics arise from microbial organisms
occupying specialized environmental niches),
and in the importance of pure basic science as
these opsins were studied over decades for
their own sake by biophysicists and
microbiologists, without involving consideration
of their potential value in delivering insights into
neuroscience and neuropsychiatric disease.[38]

Light-Activated Proteins: Channels, pumps and


enzymes

The hallmark of optogenetics therefore is


introduction of fast light-activated channels,
pumps, and enzymes that allow temporally
precise manipulation of electrical and
biochemical events while maintaining cell-type
resolution through the use of specific targeting
mechanisms. Among the microbial opsins
which can be used to investigate the function of
neural systems are the channelrhodopsins
(ChR2, ChR1, VChR1, and SFOs) to excite
neurons and anion-conducting
channelrhodopsins for light-induced inhibition.
Light-driven ion pumps are also used to inhibit
neuronal activity, e.g. halorhodopsin (NpHR),[39]
enhanced halorhodopsins (eNpHR2.0 and
eNpHR3.0, see Figure 2),[40] archaerhodopsin
(Arch), fungal opsins (Mac) and enhanced
bacteriorhodopsin (eBR).[41]

Optogenetic control of well-defined biochemical


events within behaving mammals is now also
possible. Building on prior work fusing
vertebrate opsins to specific G-protein coupled
receptors[42] a family of chimeric single-
component optogenetic tools was created that
allowed researchers to manipulate within
behaving mammals the concentration of
defined intracellular messengers such as cAMP
and IP3 in targeted cells.[43] Other biochemical
approaches to optogenetics (crucially, with
tools that displayed low activity in the dark)
followed soon thereafter, when optical control
over small GTPases and adenylyl cyclases was
achieved in cultured cells using novel strategies
from several different
laboratories.[44][45][46][47][48] This emerging
repertoire of optogenetic probes now allows
cell-type-specific and temporally precise control
of multiple axes of cellular function within intact
animals.[49]

Hardware for Light Application

Another necessary factor is hardware (e.g.


integrated fiberoptic and solid-state light
sources) to allow specific cell types, even deep
within the brain, to be controlled in freely
behaving animals. Most commonly, the latter is
now achieved using the fiberoptic-coupled
diode technology introduced in 2007,[50][51][52]
though to avoid use of implanted electrodes,
researchers have engineered ways to inscribe a
"window" made of zirconia that has been
modified to be transparent and implanted in
mice skulls, to allow optical waves to penetrate
more deeply to stimulate or inhibit individual
neurons.[53] To stimulate superficial brain areas
such as the cerebral cortex, optical fibers or
LEDs can be directly mounted to the skull of the
animal. More deeply implanted optical fibers
have been used to deliver light to deeper brain
areas. Complementary to fiber-tethered
approaches, completely wireless techniques
have been developed utilizing wirelessly
delivered power to headborne LEDs for
unhindered study of complex behaviors in freely
behaving organisms.[54]

Expression of Optogenetic Actuators

Optogenetics also necessarily includes the


development of genetic targeting strategies
such as cell-specific promoters or other
customized conditionally-active viruses, to
deliver the light-sensitive probes to specific
populations of neurons in the brain of living
animals (e.g. worms, fruit flies, mice, rats, and
monkeys). In invertebrates such as worms and
fruit flies some amount of all-trans-retinal (ATR)
is supplemented with food. A key advantage of
microbial opsins as noted above is that they are
fully functional without the addition of
exogenous co-factors in vertebrates.[52]
Technique

Three primary components in the application of


Optogenetics are as follows (A) Identification or synthesis of
a light-sensitive protein (Opsin) such as Channelrhodopsin-2
(ChR2), Halorhodopsin (NpHR), etc... (B) The design of a
system to introduce the genetic material containing the
opsin into cells for protein expression such as application of
Cre-Recombinase or an Adeno-Associated-Virus (C)
Application of light emitting instruments.[55]

The technique of using optogenetics is flexible


and adaptable to the experimenter's needs. For
starters, experimenters genetically engineer a
microbial opsin based on the gating properties
(rate of excitability, refractory period, etc..)
required for the experiment.

There is a challenge in introducing the microbial


opsin, an optogenetic actuator, into a specific
region of the organism in question. A
rudimentary approach is to introduce an
engineered viral vector that contains the
optogenetic actuator gene attached to a
recognizable promoter such as CAMKIIα. This
allows for some level of specificity as cells that
already contain and can translate the given
promoter will be infected with the viral vector
and hopefully express the optogenetic actuator
gene.
Another approach is the creation of transgenic
mice where the optogenetic actuator gene is
introduced into mice zygotes with a given
promoter, most commonly Thy1. Introduction of
the optogenetic actuator at an early stage
allows for a larger genetic code to be
incorporated and as a result, increases the
specificity of cells to be infected.

A third and rather novel approach that has been


developed is creating transgenic mice with Cre-
Recombinase, an enzyme that catalyzes
recombination between two lox-P sites. Then by
introducing an engineered viral vector
containing the optogenetic actuator gene in
between two lox-P sites, only the cells
containing the Cre-Recombinase will express
the microbial opsin. This last technique has
allowed for multiple modified optogenetic
actuators to be used without the need to create
a whole line of transgenic animals every time a
new microbial opsin is needed.

After the introduction and expression of the


microbial opsin, depending on the type of
analysis being performed, application of light
can be placed at the terminal ends or the main
region where the infected cells are situated.
Light stimulation can be performed with a vast
array of instruments from light emitting diodes
(LEDs) or diode-pumped solid state (DPSS).
These light sources are most commonly
connected to a computer through a fiber optic
cable. Recent advances include the advent of
wireless head-mounted devices that also apply
LED to targeted areas and as a result give the
animal more freedom of mobility to reproduce
in vivo results.[56][57]

Issues
Although already a powerful scientific tool,
optogenetics, according to Doug Tischer &
Orion D. Weiner of the University of California
San Francisco, should be regarded as a "first-
generation GFP" because of its immense
potential for both utilization and
optimization.[58] With that being said, the
current approach to optogenetics is limited
primarily by its versatility. Even within the field
of Neuroscience where it is most potent, the
technique is less robust on a subcellular
level.[59]
Selective expression

One of the main problems of optogenetics is


that not all the cells in question may express
the microbial opsin gene at the same level.
Thus, even illumination with a defined light
intensity will have variable effects on individual
cells. Optogenetic stimulation of neurons in the
brain is even less controlled as the light
intensity drops exponentially from the light
source (e.g. implanted optical fiber).

Moreover, mathematical modelling shows that


selective expression of opsin in specific cell
types can dramatically alter the dynamical
behavior of the neural circuitry. In particular,
optogenetic stimulation that preferentially
targets inhibitory cells can transform the
excitability of the neural tissue from Type 1 —
where neurons operate as integrators — to Type
2 where neurons operate as resonators.[60] Type
1 excitable media sustain propagating waves of
activity whereas Type 2 excitable media do not.
The transformation from one to the other
explains how constant optical stimulation of
primate motor cortex elicits gamma-band (40–
80 Hz) oscillations in the manner of a Type 2
excitable medium. Yet those same oscillations
propagate far into the surrounding tissue in the
manner of a Type 1 excitable medium.[61]

Nonetheless, it remains difficult to target opsin


to defined subcellular compartments, e.g. the
plasma membrane, synaptic vesicles, or
mitochondria.[59][62] Restricting the opsin to
specifiic regions of the plasma membrane such
as dendrites, somata or axon terminals would
provide a more robust understanding of
neuronal circuitry.[59]

Kinetics and synchronization

An issue with channelrhodopsin-2 is that its


gating properties don't mimic in vivo cation
channels of cortical neurons. A solution to this
issue with a protein's kinetic property is
introduction of variants of Channelrhodopsin-2
with more favorable kinetics.[55][56]

Another one of the technique's limitations is


that light stimulation produces a synchronous
activation of infected cells and this removes
any individual cell properties of activation
among the population affected. Therefore, it is
difficult to understand how the cells in the
population affected communicate with one
another or how their phasic properties of
activation may relate to the circuitry being
observed.

Optogenetic activation has been combined with


functional magnetic resonance imaging (ofMRI)
to elucidate the connectome, a thorough map of
the brain’s neural connections. The results,
however, are limited by the general properties of
fMRI.[59][63] The readouts from this
neuroimaging procedure lack the spatial and
temporal resolution appropriate for studying the
densely packed and rapid-firing neuronal
circuits.[63]

Excitation spectrum
The opsin proteins currently in use have
absorption peaks across the visual spectrum,
but remain considerable sensitivity to blue
light.[59] This spectral overlap makes it very
difficult to combine opsin activation with
genenetically encoded indictors (GEVIs, GECIs,
GluSnFR, synapto-pHluorin), most of which
need blue light exciation. Opsins with infrared
activation would, at a standard irradiance value,
increase light penetration and augment
resolution through reduction of light scattering.

Applications
The field of optogenetics has furthered the
fundamental scientific understanding of how
specific cell types contribute to the function of
biological tissues such as neural circuits in vivo
(see references from the scientific literature
below). Moreover, on the clinical side,
optogenetics-driven research has led to insights
into Parkinson's disease[64][65] and other
neurological and psychiatric disorders. Indeed,
optogenetics papers in 2009 have also provided
insight into neural codes relevant to autism,
Schizophrenia, drug abuse, anxiety, and
depression.[41][66][67][68]

Identification of particular neurons


and networks

Amygdala

Optogenetic approaches have been used to


map neural circuits in the amygdala that
contribute to fear conditioning.[69][70][71][72] One
such example of a neural circuit is the
connection made from the basolateral
amygdala to the dorsal-medial prefrontal cortex
where neuronal oscillations of 4 Hz have been
observed in correlation to fear induced freezing
behaviors in mice. Transgenic mice were
introduced with Channelrhodoposin-2 attached
with a Parvalbumin-Cre promoter that
selectively infected interneurons located both in
the basolateral amygdala and the dorsal-medial
prefrontal cortex responsible for the 4 Hz
oscillations. The interneurons were optically
stimulated generating a freezing behavior and
as a result provided evidence that these 4 Hz
oscillations may be responsible for the basic
fear response produced by the neuronal
populations along the dorsal-medial prefrontal
cortex and basolateral amygdala.[73]

Olfactory bulb

Optogenetic activation of olfactory sensory


neurons was critical for demonstrating timing in
odor processing[74] and for mechanism of
neuromodulatory mediated olfactory guided
behaviors (e.g. aggression, mating)[75] In
addition, with the aid of optogenetics, evidence
has been reproduced to show that the
"afterimage" of odors is concentrated more
centrally around the olfactory bulb rather than
on the periphery where the olfactory receptor
neurons would be located. Transgenic mice
infected with channel-rhodopsin Thy1-ChR2,
were stimulated with a 473 nm laser
transcranially positioned over the dorsal section
of the olfactory bulb. Longer photostimulation
of mitral cells in the olfactory bulb led to
observations of longer lasting neuronal activity
in the region after the photostimulation had
ceased, meaning the olfactory sensory system
is able to undergo long term changes and
recognize differences between old and new
odors.[76]

Nucleus accumbens

Optogenetics, freely moving mammalian


behavior, in vivo electrophysiology, and slice
physiology have been integrated to probe the
cholinergic interneurons of the nucleus
accumbens by direct excitation or inhibition.
Despite representing less than 1% of the total
population of accumbal neurons, these
cholinergic cells are able to control the activity
of the dopaminergic terminals that innervate
medium spiny neurons (MSNs) in the nucleus
accumbens.[77] These accumbal MSNs are
known to be involved in the neural pathway
through which cocaine exerts its effects,
because decreasing cocaine-induced changes
in the activity of these neurons has been shown
to inhibit cocaine conditioning. The few
cholinergic neurons present in the nucleus
accumbens may prove viable targets for
pharmacotherapy in the treatment of cocaine
dependence[41]
Cages for rat equipped of optogenetics leds commutators
which permit in vivo to study animal behavior during
optogenetics' stimulations.

Prefrontal cortex

In vivo and in vitro recordings (by the Cooper


laboratory ) of individual CAMKII AAV-ChR2
expressing pyramidal neurons within the
prefrontal cortex demonstrated high fidelity
action potential output with short pulses of blue
light at 20 Hz (Figure 1).[34] The same group
recorded complete green light-induced
silencing of spontaneous activity in the same
prefrontal cortical neuronal population
expressing an AAV-NpHR vector (Figure 2).[34]

Heart

Optogenetics was applied on atrial


cardiomyocytes to end spiral wave arrhythmias,
found to occur in atrial fibrillation, with light.[78]
This method is still in the development stage. A
recent study explored the possibilities of
optogenetics as a method to correct for
arrythmias and resynchronize cardiac pacing.
The study introduced Channelrhodopsin-2 into
cardiomyocytes in ventricular areas of hearts of
transgenic mice and performed in vitro studies
of photostimulation on both open-cavity and
closed-cavity mice. Photostimulation led to
increased activation of cells and thus increased
ventricular contractions resulting in increasing
heart rates. In addition, this approach has been
applied in cardiac resynchronization therapy
(CRT) as a new biological pacemaker as a
substitute for electrode based-CRT.[79] Lately,
optogenetics has been used in the heart to
defibrillate ventricular arrhythmias with local
epicardial illumination,[80] a generalized whole
heart illumination[81] or with customized
stimulation patterns based on arrhythmogenic
mechanisms in order to lower defibrillation
energy.[82]

Spiral ganglion

Optogenetic stimulation of the spiral ganglion in


deaf mice restored auditory activity.[83]
Optogenetic application onto the cochlear
region allows for the stimulation or inhibition of
the spiral ganglion cells (SGN). In addition, due
to the characteristics of the resting potentials
of SGN's, different variants of the protein
Channelrhodopsin-2 have been employed such
as Chronos and CatCh. Chronos and CatCh
variants are particularly useful in that they have
less time spent in their deactivated states,
which allow for more activity with less bursts of
blue light emitted. The result being that the LED
producing the light would require less energy
and the idea of cochlear prosthetics in
association with photo-stimulation, would be
more feasible.[84]

Brainstem
Optogenetic stimulation of a modified red-light
excitable channelrhodopsin (ReaChR)
expressed in the facial motor nucleus enabled
minimally invasive activation of motoneurons
effective in driving whisker movements in
mice.[85] One novel study employed
optogenetics on the Dorsal Ralphe Nucleus to
both activate and inhibit dopaminergic release
onto the Ventral Tegmental Area. To produce
activation transgenic mice were infected with
Channelrhodopsin-2 with a TH-Cre promoter
and to produce inhibition the hyperpolarizing
opsin NpHR was added onto the TH-Cre
promoter. Results showed that optically
activating dopaminergic neurons led to an
increase in social interactions, and their
inhibition decreased the need to socialize only
after a period of isolation.[86]

Precise temporal control of


interventions

The currently available optogenetic actuators


allow for the accurate temporal control of the
required intervention (i.e. inhibition or excitation
of the target neurons) with precision routinely
going down to the millisecond level. Therefore,
experiments can now be devised where the light
used for the intervention is triggered by a
particular element of behavior (to inhibit the
behavior), a particular unconditioned stimulus
(to associate something to that stimulus) or a
particular oscillatory event in the brain (to
inhibit the event). This kind of approach has
already been used in several brain regions:

Hippocampus

Sharp waves and ripple complexes (SWRs) are


distinct high frequency oscillatory events in the
hippocampus thought to play a role in memory
formation and consolidation. These events can
be readily detected by following the oscillatory
cycles of the on-line recorded local field
potential. In this way the onset of the event can
be used as a trigger signal for a light flash that
is guided back into the hippocampus to inhibit
neurons specifically during the SWRs and also
to optogenetically inhibit the oscillation itself[87]
These kinds of "closed-loop" experiments are
useful to study SWR complexes and their role in
memory.

Cellular biology/cell signaling


pathways

Optogenetic control of cellular forces and induction of


mechanotransduction. Pictured cells receive an hour of
imaging concurrent with blue light that pulses every 60
seconds. This is also indicated when the blue point flashes
onto the image. The cell relaxes for an hour without light
activation and then this cycle repeats again. The square inset
magnifies the cell's nucleus.

The optogenetic toolkit has proven pivotal for


the field of neuroscience as it allows precise
manipulation of neuronal excitability. Moreover,
this technique has been shown to extend
outside neurons to an increasing number of
proteins and cellular functions.[58] Cellular scale
modifications including manipulation of
contractile forces relevant to cell migration, cell
division and wound healing have been
optogenetically manipulated.[88] The field has
not developed to the point where processes
crucial to cellular and developmental biology
and cell signaling including protein localization,
post-translational modification and GTP loading
can be consistently controlled via
optogenetics.[58]

Photosensitive proteins utilized in


various cell signaling pathways

While this extension of optogenetics remains to


be further investigated, there are various
conceptual methodologies that may prove to
immediately robust. There is a considerable
body of literature outlining photosensitive
proteins that have been utilized in cell signaling
pathways.[58] CRY2, LOV, DRONPA and PHYB are
photosynthetic proteins involved in inducible
protein association whereby activation via light
can induce/turn off a signaling cascade via
recruitment of a signaling domain to its
respective substrate.[89][90][91][92] LOV and PHYB
are photosensitive proteins that engage in
homodimerization and/or heterodimerization to
recruit some DNA-modifying protein,
translocate to the site of DNA and alter gene
expression levels.[93][94][95] CRY2, a protein that
inherently clusters when active, has been fused
with signaling domains and subsequently
photoactivated allowing for clustering-based
activation.[96] Proteins LOV and Dronpa have
also been adapted to cell signaling
manipulation; exposure to light induces
conformational changes in the photosensitive
protein which can subsequently reveal a
previously obscured signaling domain and/or
activate a protein that was otherwise
allosterically inhibited.[97][98] LOV has been
fused to caspase 3 to produce a construct
capable of inducing apoptosis upon light
stimulation.[99]

Optogenetic temporal control of


signals

A different set of signaling cascades respond to


stimulus timing duration and dynamics.[100]
Adaptive signaling pathways, for instance,
adjust in accordance to the current level of the
projected stimulus and display activity only
when these levels change as opposed to
responding to absolute levels of the input.[101]
Stimulus dynamics also can trigger activity;
treating PC12 cells with epidermal growth
factor (inducing a transient profile of ERK
activity) leads to cellular proliferation whereas
introduction of nerve growth factor (inducing a
sustained profile of ERK activity) is associated
with a different cellular decision whereby the
PC12 cells differentiate into neuron-like
cells.[102] This discovery was guided
pharmacologically but the finding was
replicated utilizing optogenetic inputs
instead.[103] This ability to optogenetically
control signals for various time durations is
being explored to elucidate various cell
signaling pathways where there is not a strong
enough understanding to utilize either
drug/genetic manipulation.[58]

References
1. Deisseroth, K.; Feng, G.; Majewska, A. K.;
Miesenbock, G.; Ting, A.; Schnitzer, M. J. (2006).
"Next-Generation Optical Technologies for
Illuminating Genetically Targeted Brain Circuits" .
Journal of Neuroscience. 26 (41): 10380–6.
doi:10.1523/JNEUROSCI.3863-06.2006 .
PMC 2820367  . PMID 17035522 .
2. Mancuso, J. J.; Kim, J.; Lee, S.; Tsuda, S.;
Chow, N. B. H.; Augustine, G. J. (2010).
"Optogenetic probing of functional brain
circuitry". Experimental Physiology. 96 (1): 26–
33. doi:10.1113/expphysiol.2010.055731 .
PMID 21056968 .
3. Treger, Jeremy (2015). "Single-molecule
fluorimetry and gating currents inspire an
improved optical voltage indicator" . eLife. 4:
e10482. doi:10.7554/eLife.10482 .
PMC 4658195  . PMID 26599732 .
4. Primer on Optogenetics: Pastrana, Erika
(2010). "Optogenetics: Controlling cell function
with light" . Nature Methods. 8 (1): 24–25.
doi:10.1038/nmeth.f.323 .
Editorial: "Method of the Year 2010" . Nature
Methods. 8 (1): 1. 2010.
doi:10.1038/nmeth.f.321 .
Commentary: Deisseroth, Karl (2010).
"Optogenetics" . Nature Methods. 8 (1): 26–9.
doi:10.1038/nmeth.f.324 . PMID 21191368 .
5. News, Staff (2010). "Insights of the decade.
Stepping away from the trees for a look at the
forest. Introduction". Science. 330 (6011): 1612–
3. Bibcode:2010Sci...330.1612. .
doi:10.1126/science.330.6011.1612 .
PMID 21163985 .
6. Crick, F. (December 1999). "The impact of
molecular biology on neuroscience" .
Philosophical Transactions of the Royal Society
B. 354 (1392): 2021–25.
doi:10.1098/rstb.1999.0541 . PMC 1692710  .
PMID 10670022 .
7. Fork, R. L. (March 1971). "Laser stimulation of
nerve cells in Aplysia" . Science. 171 (3974):
907–8. Bibcode:1971Sci...171..907F .
doi:10.1126/science.171.3974.907 .
PMID 5541653 .
8. Zemelman, B. V.; Lee, G. A.; Ng, M.;
Miesenböck, G. (2002). "Selective
photostimulation of genetically chARGed
neurons". Neuron. 33 (1): 15–22.
doi:10.1016/S0896-6273(01)00574-8 .
PMID 11779476 .
9. Zemelman, B. V.; Nesnas, N.; Lee, G.A.;
Miesenböck, G. (2003). "Photochemical gating of
heterologous ion channels: Remote control over
genetically designated populations of neurons" .
PNAS. 100: 1352–7.
doi:10.1073/pnas.242738899 . PMC 298776  .
PMID 12540832 .
10. Banghart, M; Borges, K; Isacoff, E; Trauner, R.
H. (21 November 2004). "Light-activated ion
channels for remote control of neuronal firing" .
Nature Neuroscience. 7 (12): 1381–1386.
doi:10.1038/nn1356 . PMC 1447674  .
PMID 15558062 .
11. Volgraf,, M.; Gorostiza, P.; Numano, R.;
Kramer, R. H.; Isacoff, E. Y. (11 December 2005).
"Allosteric control of an ionotropic glutamate
receptor with an optical switch" . Nature
Chemical Biology. 2 (1): 47–52.
doi:10.1038/nchembio756 . PMC 1447676  .
PMID 16408092 .
12. Arenkiel, BR; Klein, ME; Davison, IG; Katz, LC;
Ehlers, MD (2008). "Genetic control of neuronal
activity in mice conditionally expressing
TRPV1" . Nature Methods. 5: 299–302.
doi:10.1038/nmeth.1190 . PMC 3127246  .
PMID 18327266 . Retrieved 2017-03-15.
13. "Transient activation of specific neurons in
mice by selective expression of the capsaicin
receptor" . Retrieved 2017-03-15.
14. "Synaptic Modifications in the Medial
Prefrontal Cortex in Susceptibility and Resilience
to Stress" . JNeurosci. Retrieved 2017-03-15.
15. Nagel,, G.; Szellas, T.; Huhn, W.; Kateriya, S.;
Adeishvili, N.; Berthold, P.; Ollig, D.; Hegemann, P.;
Bamberg, E. (25 November 2003).
"Channelrhodopsin-2, a directly light-gated
cation-selective membrane channel" . Proc Natl
Acad Sci U S A. 100 (24): 13940–5.
Bibcode:2003PNAS..10013940N .
doi:10.1073/pnas.1936192100 . PMC 283525  .
PMID 14615590 .
16. Harz, Hartmann; Hegemann, Peter (1991-06-
06). "Rhodopsin-regulated calcium currents in
Chlamydomonas" . Nature. 351 (6326): 489–
491. doi:10.1038/351489a0 .
17. Nagel, Georg; Ollig, Doris; Fuhrmann, Markus;
Kateriya, Suneel; Musti, Anna Maria; Bamberg,
Ernst; Hegemann, Peter (2002-06-28).
"Channelrhodopsin-1: A Light-Gated Proton
Channel in Green Algae" . Science. 296 (5577):
2395–2398. doi:10.1126/science.1072068 .
ISSN 0036-8075 . PMID 12089443 .
18. Bi, Anding; Cui, Jinjuan; Ma, Yu-Ping;
Olshevskaya, Elena; Pu, Mingliang; Dizhoor,
Alexander M.; Pan, Zhuo-Hua (2006). "Ectopic
Expression of a Microbial-Type Rhodopsin
Restores Visual Responses in Mice with
Photoreceptor Degeneration" . Neuron. 50 (1):
23–33. doi:10.1016/j.neuron.2006.02.026 .
ISSN 0896-6273 . PMC 1459045  .
PMID 16600853 .
19. Lima, S. Q.; Miesenböck, G. (2005). "Remote
Control of Behavior through Genetically Targeted
Photostimulation of Neurons". Cell. 121 (1):
141–152. doi:10.1016/j.cell.2005.02.004 .
PMID 15820685 .
20. Boyden, E. S.; Zhang, F.; Bamberg, E.; Nagel,
G.; Deisseroth, K. (2005). "Millisecond-timescale,
genetically targeted optical control of neural
activity". Nat. Neurosci. 8 (9): 1263–8.
doi:10.1038/nn1525 . PMID 16116447 .
21. Li,, X.; Gutierrez, D. V.; Hanson, M. G.; Han, J.;
Mark, M. D.; Chiel, H.; Hegemann, P.; Landmesser,
L. T.; Herlitze, S. (14 October 2005). "Fast
noninvasive activation and inhibition of neural
and network activity by vertebrate rhodopsin and
green algae channelrhodopsin" (PDF). Proc Natl
Acad Sci U S A. 102 (49): 17816–21.
Bibcode:2005PNAS..10217816L .
doi:10.1073/pnas.0509030102 . PMC 1292990  .
PMID 16306259 .
22. Nagel, G.; Brauner, M.; Liewald, J. F.;
Adeishvili, N.; Bamberg, E.; Gottschalk, A.
(December 2005). "Light activation of
channelrhodopsin-2 in excitable cells of
Caenorhabditis elegans triggers rapid behavioral
responses". Curr. Biol. 15 (24): 2279–84.
doi:10.1016/j.cub.2005.11.032 .
PMID 16360690 .
23. Tsien, JZ; et al. (Dec 1996). "Subregion- and
cell type-restricted gene knockout in mouse
brain" . Cell. 87 (7): 1317–26.
doi:10.1016/S0092-8674(00)81826-7 .
PMID 8980237 .
24. Tsien, JZ (2016). "Cre-Lox Neurogenetics: 20
Years of Versatile Applications in Brain Research
and Counting" . Front Genet. 7: 19.
doi:10.3389/fgene.2016.00019 . PMC 4759636 
. PMID 26925095 .
25. Miyawaki, A.; Llopis, J.; Heim, R.; McCaffery,
J. M.; Adams, J. A.; Ikura, M.; Tsien, R. Y. (1997-
08-28). "Fluorescent indicators for Ca2+ based
on green fluorescent proteins and calmodulin".
Nature. 388 (6645): 882–887.
doi:10.1038/42264 . ISSN 0028-0836 .
PMID 9278050 .
26. Kerr, R.; Lev-Ram, V.; Baird, G.; Vincent, P.;
Tsien, R. Y.; Schafer, W. R. (2000-06-01). "Optical
imaging of calcium transients in neurons and
pharyngeal muscle of C. elegans". Neuron. 26
(3): 583–594. doi:10.1016/s0896-
6273(00)81196-4 . ISSN 0896-6273 .
PMID 10896155 .
27. Fiala, André; Spall, Thomas; Diegelmann,
Sören; Eisermann, Beate; Sachse, Silke; Devaud,
Jean-Marc; Buchner, Erich; Galizia, C. Giovanni
(2002-10-29). "Genetically expressed cameleon
in Drosophila melanogaster is used to visualize
olfactory information in projection neurons".
Current Biology. 12 (21): 1877–1884.
doi:10.1016/s0960-9822(02)01239-3 .
ISSN 0960-9822 . PMID 12419190 .
28. Higashijima, Shin-ichi; Masino, Mark A.;
Mandel, Gail; Fetcho, Joseph R. (2003-12-01).
"Imaging neuronal activity during zebrafish
behavior with a genetically encoded calcium
indicator". Journal of Neurophysiology. 90 (6):
3986–3997. doi:10.1152/jn.00576.2003 .
ISSN 0022-3077 . PMID 12930818 .
29. Ji, Guangju; Feldman, Morris E.; Deng, Ke-Yu;
Greene, Kai Su; Wilson, Jason; Lee, Jane C.;
Johnston, Robyn C.; Rishniw, Mark; Tallini,
Yvonne (2004-05-14). "Ca2+-sensing transgenic
mice: postsynaptic signaling in smooth muscle".
The Journal of Biological Chemistry. 279 (20):
21461–21468. doi:10.1074/jbc.M401084200 .
ISSN 0021-9258 . PMID 14990564 .
30. Nakai, J.; Ohkura, M.; Imoto, K. (2001-02-01).
"A high signal-to-noise Ca(2+) probe composed
of a single green fluorescent protein". Nature
Biotechnology. 19 (2): 137–141.
doi:10.1038/84397 . ISSN 1087-0156 .
PMID 11175727 .
31. Chen, Tsai-Wen; Wardill, Trevor J.; Sun, Yi;
Pulver, Stefan R.; Renninger, Sabine L.; Baohan,
Amy; Schreiter, Eric R.; Kerr, Rex A.; Orger,
Michael B. (2013-07-18). "Ultrasensitive
fluorescent proteins for imaging neuronal
activity" . Nature. 499 (7458): 295–300.
doi:10.1038/nature12354 . ISSN 1476-4687 .
PMC 3777791  . PMID 23868258 .
32. "The Brain Prize 2013" . Retrieved 3 October
2013.
33. Reiner, A.; Isacoff, E.Y. (October 2013). "The
Brain Prize 2013: the optogenetics revolution".
Trends Neurosci. 36 (10): 557–60.
doi:10.1016/j.tins.2013.08.005 .
PMID 24054067 .
34. Baratta M.V., Nakamura S, Dobelis P.,
Pomrenze M.B., Dolzani S.D. & Cooper D.C.
(2012) Optogenetic control of genetically-
targeted pyramidal neuron activity in prefrontal
cortex. Nature Precedings April 2
doi=10.1038/npre.2012.7102.1
http://www.neuro-cloud.net/nature-
precedings/baratta
35. Husson, S. J.; Liewald, J. F.; Schultheis, C.;
Stirman, J. N.; Lu, H.; Gottschalk, A. (2012).
Samuel, Aravinthan, ed. "Microbial Light-
Activatable Proton Pumps as Neuronal Inhibitors
to Functionally Dissect Neuronal Networks in C.
Elegans" . PLoS ONE. 7 (7): e40937.
doi:10.1371/journal.pone.0040937 .
PMC 3397962  . PMID 22815873 .
36. Liu, Y.; Lebeouf, B.; Guo, X.; Correa, P. A.;
Gualberto, D. G.; Lints, R.; Garcia, L. R. (2011).
Goodman, Miriam B, ed. "A Cholinergic-
Regulated Circuit Coordinates the Maintenance
and Bi-Stable States of a Sensory-Motor
Behavior during Caenorhabditis elegans Male
Copulation" . PLoS Genetics. 7 (3): e1001326.
doi:10.1371/journal.pgen.1001326 .
PMC 3053324  . PMID 21423722 .
37. Akemann, W; Mutoh, H; Perron, A; Park, YK;
Iwamoto, Y; Knöpfel, T (2012). "Imaging neural
circuit dynamics with a voltage-sensitive
fluorescent protein" . J Neurophysiol. 108 (8):
2323–37. doi:10.1152/jn.00452.2012 .
PMID 22815406 .
38. Deisseroth, Karl. "Optogenetics: Controlling
the Brain with Light [Extended Version]" .
Scientific American. Retrieved 2016-11-28.
39. Zhao, S.; Cunha, C.; Zhang, F.; Liu, Q.; Gloss,
B.; Deisseroth, K.; Augustine, G. J.; Feng, G.
(2008). "Improved expression of halorhodopsin
for light-induced silencing of neuronal activity" .
Brain Cell Biology. 36 (1–4): 141–154.
doi:10.1007/s11068-008-9034-7 .
PMC 3057022  . PMID 18931914 .
40. Gradinaru, V.; Thompson, K. R.; Deisseroth, K.
(2008). "ENpHR: A Natronomonas halorhodopsin
enhanced for optogenetic applications" . Brain
Cell Biology. 36 (1–4): 129–139.
doi:10.1007/s11068-008-9027-6 .
PMC 2588488  . PMID 18677566 .
41. Witten, I. B.; Lin, S. C.; Brodsky, M.; Prakash,
R.; Diester, I.; Anikeeva, P.; Gradinaru, V.;
Ramakrishnan, C.; Deisseroth, K. (2010).
"Cholinergic interneurons control local circuit
activity and cocaine conditioning" . Science. 330
(6011): 1677–81.
doi:10.1126/science.1193771 . PMC 3142356  .
PMID 21164015 .
42. Kim, J. M.; Hwa, J.; Garriga, P.; Reeves, P. J.;
RajBhandary, U. L.; Khorana, H. G. (2005). "Light-
driven activation of beta 2-adrenergic receptor
signaling by a chimeric rhodopsin containing the
beta 2-adrenergic receptor cytoplasmic loops".
Biochemistry. 44 (7): 2284–92.
doi:10.1021/bi048328i . PMID 15709741 .
43. Airan, R. D.; Thompson, K. R.; Fenno, L. E.;
Bernstein, H.; Deisseroth, K. (2009). "Temporally
precise in vivo control of intracellular signalling".
Nature. 458 (7241): 1025–9.
Bibcode:2009Natur.458.1025A .
doi:10.1038/nature07926 . PMID 19295515 .
44. Levskaya, Anselm; Weiner, Orion D.; Lim,
Wendell A.; Voigt, Christopher A. (October 2009).
"Spatiotemporal control of cell signalling using a
light-switchable protein interaction" . Nature.
461 (7266): 997–1001.
Bibcode:2009Natur.461..997L .
doi:10.1038/nature08446 . PMC 2989900  .
PMID 19749742 .
45. Wu, Yi I.; Frey, Daniel; Lungu, Oana I.; Jaehrig,
Angelika; Schlichting, Ilme; Kuhlman, Brian;
Hahn, Klaus M. (September 2009). "A genetically
encoded photoactivatable Rac controls the
motility of living cells" . Nature. 461 (7260):
104–8. Bibcode:2009Natur.461..104W .
doi:10.1038/nature08241 . PMC 2766670  .
PMID 19693014 .
46. Yazawa, M.; Sadaghiani, A. M.; Hsueh, B.;
Dolmetsch, R. E. (2009). "Induction of protein-
protein interactions in live cells using light".
Nature Biotechnology. 27 (10): 941–5.
doi:10.1038/nbt.1569 . PMID 19801976 .
47. Stierl, M.; Stumpf, P.; Udwari, D.; Gueta, R.;
Hagedorn, R.; Losi, A.; Gartner, W.; Petereit, L.; et
al. (January 2011). "Light modulation of cellular
cAMP by a small bacterial photoactivated
adenylyl cyclase, bPAC, of the soil bacterium
Beggiatoa" . J. Biol. Chem. 286 (2): 1181–8.
doi:10.1074/jbc.M110.185496 . PMC 3020725  .
PMID 21030594 .
48. Ryu, M.-H.; Moskvin, O. V.; Siltberg-Liberles,
J.; Gomelsky, M. (December 2010). "Natural and
engineered photoactivated nucleotidyl cyclases
for optogenetic applications" . J. Biol. Chem.
285 (53): 41501–8.
doi:10.1074/jbc.M110.177600 . PMC 3009876  .
PMID 21030591 .
49. Lerner, TN; Ye, L; Deisseroth, K (2016).
"Communication in Neural Circuits: Tools,
Opportunities, and Challenges" . Cell. 164 (6):
1136–50. doi:10.1016/j.cell.2016.02.027 .
PMID 26967281 .
50. Aravanis, Alexander M; Wang, Li-Ping; Zhang,
Feng; Meltzer, Leslie A; Mogri, Murtaza Z;
Schneider, M Bret; Deisseroth, Karl (September
2007). "An optical neural interface: in vivo control
of rodent motor cortex with integrated fiberoptic
and optogenetic technology". J Neural Eng. 4 (3):
S143–56. Bibcode:2007JNEng...4S.143A .
doi:10.1088/1741-2560/4/3/S02 .
PMID 17873414 .. PMID 17873414
51. Adamantidis, Antoine R.; Zhang, Feng;
Aravanis, Alexander M.; Deisseroth, Karl; De
Lecea, Luis (November 2007). "Neural substrates
of awakening probed with optogenetic control of
hypocretin neurons". Nature. 450 (7168): 420–4.
Bibcode:2007Natur.450..420A .
doi:10.1038/nature06310 . PMID 17943086 .
52. Gradinaru, V.; Thompson, K. R.; Zhang, F.;
Mogri, M.; Kay, K.; Schneider, M. B.; Deisseroth, K.
(2007). "Targeting and readout strategies for fast
optical neural control in vitro and in vivo". J.
Neurosci. 27 (52): 14231–8.
doi:10.1523/JNEUROSCI.3578-07.2007 .
PMID 18160630 .
53. Damestani, Yasaman; Reynolds, Carissa L.;
Szu, Jenny; Hsu, Mike S.; Kodera, Yasuhiro;
Binder, Devin K.; Park, B. Hyle; Garay, Javier E.;
Rao, Masaru P.; Aguilar, Guillermo (2013).
"Transparent nanocrystalline yttria-stabilized-
zirconia calvarium prosthesis" . Nanomedicine.
Elsevier Inc. 9 (8): 1135–8.
doi:10.1016/j.nano.2013.08.002 .
PMID 23969102 . Retrieved September 11, 2013.
• Explained by Mohan, Geoffrey (September 4,
2013). "A window to the brain? It's here, says UC
Riverside team" . Los Angeles Times.
54. Wentz, Christian T; Bernstein, Jacob G;
Monahan, Patrick; Guerra, Alexander; Rodriguez,
Alex; Boyden, Edward S (2011). "A wirelessly
powered and controlled device for optical neural
control of freely-behaving animals" . Journal of
Neural Engineering. 8 (4): 046021.
Bibcode:2011JNEng...8d6021W .
doi:10.1088/1741-2560/8/4/046021 .
PMC 3151576  . PMID 21701058 .
55. Pama, E. A. Claudia; Colzato, Lorenza S.;
Hommel, Bernhard (2013-01-01). "Optogenetics
as a neuromodulation tool in cognitive
neuroscience" . Cognition. 4: 610.
doi:10.3389/fpsyg.2013.00610 . PMC 3764402  .
PMID 24046763 .
56. Warden, Melissa R.; Cardin, Jessica A.;
Deisseroth, Karl (2014-07-11). "Optical Neural
Interfaces" . Annual review of biomedical
engineering. 16: 103–129. doi:10.1146/annurev-
bioeng-071813-104733 . ISSN 1523-9829 .
PMC 4163158  . PMID 25014785 .
57. Guru, Akash; Post, Ryan J; Ho, Yi-Yun;
Warden, Melissa R (2015-07-25). "Making Sense
of Optogenetics" . International Journal of
Neuropsychopharmacology. 18 (11): pyv079.
doi:10.1093/ijnp/pyv079 . ISSN 1461-1457 .
PMC 4756725  . PMID 26209858 .
58. Tischer, Doug; Weiner, Orion D. (2014-08-01).
"Illuminating cell signalling with optogenetic
tools" . Nature Reviews Molecular Cell Biology.
15 (8): 551–558. doi:10.1038/nrm3837 .
ISSN 1471-0072 . PMC 4145075  .
PMID 25027655 .
59. Zalocusky, Kelly A; Fenno, Lief E; Deisseroth,
Karl (2013). "Current Challenges in
Optogenetics" . Society for Neuroscience.
60. Heitmann, Stewart; Rule, Michael; Truccolo,
Wilson; Ermentrout, Bard (2017). "Optogenetic
stimulation shifts the excitability of cerebral
cortex from Type I to Type II: Oscillation onset
and wave propagation". PLOS Computational
Biology. 13 (1): e1005349.
doi:10.1371/journal.pcbi.1005349 . ISSN 1553-
7358 .
61. Lu, Yao; Truccolo, Wilson; Wagner, Fabien B.;
Vargas-Irwin, Carlos E.; Ozden, Ilker;
Zimmermann, Jonas B.; May, Travis; Agha,
Naubahar S.; Wang, Jing; Nurmikko, Arto V.
(2016). "Optogenetically induced spatiotemporal
gamma oscillations and neuronal spiking activity
in primate motor cortex". Journal of
Neurophysiology. 113 (10): 3574–3587.
doi:10.1152/jn.00792.2014 . ISSN 1522-1598 .
62. Gradinaru, Viviana; Thompson, Kimberly R.;
Deisseroth, Karl (2008-08-01). "eNpHR: a
Natronomonas halorhodopsin enhanced for
optogenetic applications" . Brain Cell Biology. 36
(1-4): 129–139. doi:10.1007/s11068-008-9027-
6 . ISSN 1559-7113 . PMC 2588488  .
PMID 18677566 .
63. Leergaard, Trygve B.; Hilgetag, Claus C.;
Sporns, Olaf (2012-05-01). "Mapping the
Connectome: Multi-Level Analysis of Brain
Connectivity" . Frontiers in Neuroinformatics. 6.
doi:10.3389/fninf.2012.00014 . ISSN 1662-
5196 . PMC 3340894  . PMID 22557964 .
64. Kravitz, A. V.; Freeze, B. S.; Parker, P. R. L.;
Kay, K.; Thwin, M. T.; Deisseroth, K.; Kreitzer, A. C.
(2010). "Regulation of parkinsonian motor
behaviours by optogenetic control of basal
ganglia circuitry" . Nature. 466 (7306): 622–626.
doi:10.1038/nature09159 . PMC 3552484  .
PMID 20613723 .
65. Gradinaru, V.; Mogri, M.; Thompson, K. R.;
Henderson, J. M.; Deisseroth, K. (2009). "Optical
Deconstruction of Parkinsonian Neural Circuitry".
Science. 324 (5925): 354–359.
doi:10.1126/science.1167093 .
PMID 19299587 .
66. Cardin, J. A.; Carlén, M.; Meletis, K.; Knoblich,
Ulf; Zhang, Feng; Deisseroth, Karl; Tsai, Li-Huei;
Moore, Christopher I. (2009). "(June 2009).
"Driving fast-spiking cells induces gamma
rhythm and controls sensory responses" .
Nature. 459 (7247): 663–7.
Bibcode:2009Natur.459..663C .
doi:10.1038/nature08002 . PMC 3655711  .
PMID 19396156 .
67. Sohal, V. S.; Zhang, F.; Yizhar, O.; Deisseroth,
K. (2009). "Parvalbumin neurons and gamma
rhythms enhance cortical circuit performance".
Nature. 459 (7247): 698–702.
Bibcode:2009Natur.459..698S .
doi:10.1038/nature07991 . PMID 19396159 .
68. Tsai, H.C.; Zhang, F.; Adamantidis, A.; Stuber,
G. D.; Bonci, A.; De Lecea, L.; Deisseroth, K.
(2009). "Phasic firing in dopaminergic neurons is
sufficient for behavioral conditioning". Science.
324 (5930): 1080–4.
Bibcode:2009Sci...324.1080T .
doi:10.1126/science.1168878 .
PMID 19389999 .
69. Haubensak, W.; Kunwar, P. S.; Cai, H.; Ciocchi,
S.; Wall, N. R.; Ponnusamy, R.; Biag, J.; Dong, H.
W.; Deisseroth, K.; Callaway, E. M.; Fanselow, M.
S.; Lüthi, A.; Anderson, D. J. (2010). "Genetic
dissection of an amygdala microcircuit that
gates conditioned fear" . Nature. 468 (7321):
270–276. doi:10.1038/nature09553 .
PMC 3597095  . PMID 21068836 .
70. Johansen, J. P.; Hamanaka, H.; Monfils, M.
H.; Behnia, R.; Deisseroth, K.; Blair, H. T.; Ledoux,
J. E. (2010). "Optical activation of lateral
amygdala pyramidal cells instructs associative
fear learning" . Proceedings of the National
Academy of Sciences. 107 (28): 12692–12697.
doi:10.1073/pnas.1002418107 . PMC 2906568  .
PMID 20615999 .
71. Jasnow AM, Ehrlich DE, Choi DC, Dabrowska
J, Bowers ME, McCullough KM, Rainnie DG,
Ressler KJ (2013). "Thy1-expressing neurons in
the basolateral amygdala may mediate fear
inhibition" . J. Neurosci. 33 (25): 10396–404.
doi:10.1523/JNEUROSCI.5539-12.2013 .
PMC 3685835  . PMID 23785152 .
72. Dias, B. G.; Banerjee, S. B.; Goodman, J. V.;
Ressler, K. J. (2013). "Towards new approaches
to disorders of fear and anxiety" . Current
Opinion in Neurobiology. 23 (3): 346–352.
doi:10.1016/j.conb.2013.01.013 . PMC 3672317 
. PMID 23402950 .
73. Karalis, Nikolaos; Dejean, Cyril; Chaudun,
Fabrice; Khoder, Suzana; Rozeske, Robert R.;
Wurtz, Hélène; Bagur, Sophie; Benchenane,
Karim; Sirota, Anton (2016-04-01). "4-Hz
oscillations synchronize prefrontal-amygdala
circuits during fear behavior" . Nature
Neuroscience. 19 (4): 605–612.
doi:10.1038/nn.4251 . ISSN 1546-1726 .
PMC 4843971  . PMID 26878674 .
74. Shusterman, R; Smear, MC; Koulakov, AA;
Rinberg, D (17 July 2011). "Precise olfactory
responses tile the sniff cycle". Nature
Neuroscience. 14 (8): 1039–44.
doi:10.1038/nn.2877 . PMID 21765422 .
75. Smith RS, Hu R, DeSouza A, Eberly CL, Krahe
K, Chan W, Araneda RC (Jul 2015). "Differential
Muscarinic Modulation in the Olfactory Bulb" .
The Journal of Neuroscience. 35 (30): 10773–
85. doi:10.1523/JNEUROSCI.0099-15.2015 .
PMC 4518052  . PMID 26224860 .
76. Patterson, Michael Andrew; Lagier, Samuel;
Carleton, Alan (2013-08-27). "Odor
representations in the olfactory bulb evolve after
the first breath and persist as an odor
afterimage" . Proceedings of the National
Academy of Sciences. 110 (35): E3340–E3349.
doi:10.1073/pnas.1303873110 . ISSN 0027-
8424 . PMC 3761593  . PMID 23918364 .
77. Tecuapetla, F.; Patel, J. C.; Xenias, H.; English,
D.; Tadros, I.; Shah, F.; Berlin, J.; Deisseroth, K.;
Rice, M. E.; Tepper, J. M.; Koos, T. (2010).
"Glutamatergic Signaling by Mesolimbic
Dopamine Neurons in the Nucleus Accumbens".
Journal of Neuroscience. 30 (20): 7105–7110.
doi:10.1523/JNEUROSCI.0265-10.2010 .
PMID 20484653 .
78. Bingen BO, Engels MC, Schalij MJ,
Jangsangthong W, Neshati Z, Feola I, et al.
(2014). "Light-induced termination of spiral wave
arrhythmias by optogenetic engineering of atrial
cardiomyocytes" . Cardiovasc Res. 104 (1): 194–
205. doi:10.1093/cvr/cvu179 . PMID 25082848 .
79. Nussinovitch, Udi; Gepstein, Lior.
"Optogenetics for in vivo cardiac pacing and
resynchronization therapies" . Nature
Biotechnology. 33 (7): 750–754.
doi:10.1038/nbt.3268 . PMID 26098449 .
80. Nyns EC, et al. (2016). "Optogenetic
termination of ventricular arrhythmias in the
whole heart: towards biological cardiac rhythm
management". Eur Heart J: ehw574.
doi:10.1093/eurheartj/ehw574 .
81. Bruegmann T, et al. (2016). "Optogenetic
defibrillation terminates ventricular arrhythmia in
mouse hearts and human simulations". J Clin
Invest. 126: 3894–3904. doi:10.1172/JCI88950 .
82. Crocini C, et al. (2016). "Optogenetics design
of mechanistically-based stimulation patterns
for cardiac defibrillation" . Sci Rep. 6.
doi:10.1038/srep35628 .
83. Hernandez, Victor H.; et al. (2014).
"Optogenetic stimulation of the auditory
pathway" . J Clin Invest. 124 (3): 1114–1129.
doi:10.1172/JCI69050 . PMC 3934189  .
PMID 24509078 .
84. Moser, Tobias. "Optogenetic stimulation of
the auditory pathway for research and future
prosthetics" . Current Opinion in Neurobiology.
34: 29–36. doi:10.1016/j.conb.2015.01.004 .
PMID 25637880 .
85. Lin, JY; Knutsen, PM; Muller, A; Kleinfeld, D;
Tsien, RY (2013). "ReaChR: A red-shifted variant
of channelrhodopsin enables deep transcranial
optogenetic excitation" . Nature Neuroscience.
16 (10): 1499–1508. doi:10.1038/nn.3502 .
PMC 3793847  . PMID 23995068 .
86. Matthews, Gillian A.; Nieh, Edward H.; Vander
Weele, Caitlin M.; Halbert, Sarah A.; Pradhan,
Roma V.; Yosafat, Ariella S.; Glober, Gordon F.;
Izadmehr, Ehsan M.; Thomas, Rain E. (2016-02-
11). "Dorsal Raphe Dopamine Neurons
Represent the Experience of Social Isolation" .
Cell. 164 (4): 617–631.
doi:10.1016/j.cell.2015.12.040 . ISSN 1097-
4172 . PMC 4752823  . PMID 26871628 .
87. Kovacs KA, O'Neill J, Schoenenberger P,
Penttonen M, Ranguel Guerrero DK, Csicsvari J
(19 Nov 2016). "Optogenetically Blocking Sharp
Wave Ripple Events in Sleep Does Not Interfere
with the Formation of Stable Spatial
Representation in the CA1 Area of the
Hippocampus" . PLOS ONE. 11: e0164675.
doi:10.1371/journal.pone.0164675 .
PMC 5070819  . PMID 27760158 .
88. Valon, Léo; Marín-Llauradó, Ariadna; Wyatt,
Thomas; Charras, Guillaume; Trepat, Xavier
(2017-02-10). "Optogenetic control of cellular
forces and mechanotransduction" . Nature
Communications. 8.
doi:10.1038/ncomms14396 . ISSN 2041-1723 .
PMC 5309899  . PMID 28186127 .
89. Strickland, Devin; Lin, Yuan; Wagner,
Elizabeth; Hope, C. Matthew; Zayner, Josiah;
Antoniou, Chloe; Sosnick, Tobin R.; Weiss, Eric L.;
Glotzer, Michael (2012-03-04). "TULIPs: tunable,
light-controlled interacting protein tags for cell
biology" . Nature Methods. 9 (4): 379–384.
doi:10.1038/nmeth.1904 . ISSN 1548-7105 .
PMC 3444151  . PMID 22388287 .
90. Idevall-Hagren, Olof; Dickson, Eamonn J.;
Hille, Bertil; Toomre, Derek K.; De Camilli, Pietro
(2012-08-28). "Optogenetic control of
phosphoinositide metabolism" . Proceedings of
the National Academy of Sciences of the United
States of America. 109 (35): E2316–2323.
doi:10.1073/pnas.1211305109 . ISSN 1091-
6490 . PMC 3435206  . PMID 22847441 .
91. Leung, Daisy W.; Otomo, Chinatsu; Chory,
Joanne; Rosen, Michael K. (2008-09-02).
"Genetically encoded photoswitching of actin
assembly through the Cdc42-WASP-Arp2/3
complex pathway" . Proceedings of the National
Academy of Sciences of the United States of
America. 105 (35): 12797–12802.
doi:10.1073/pnas.0801232105 . ISSN 1091-
6490 . PMC 2525560  . PMID 18728185 .
92. Toettcher, Jared E.; Gong, Delquin; Lim,
Wendell A.; Weiner, Orion D. (2011-09-11). "Light-
based feedback for controlling intracellular
signaling dynamics" . Nature Methods. 8 (10):
837–839. doi:10.1038/nmeth.1700 . ISSN 1548-
7105 . PMC 3184382  . PMID 21909100 .
93. Lungu, Oana I.; Hallett, Ryan A.; Choi, Eun
Jung; Aiken, Mary J.; Hahn, Klaus M.; Kuhlman,
Brian (2012-04-20). "Designing photoswitchable
peptides using the AsLOV2 domain" . Chemistry
& Biology. 19 (4): 507–517.
doi:10.1016/j.chembiol.2012.02.006 .
ISSN 1879-1301 . PMC 3334866  .
PMID 22520757 .
94. Konermann, Silvana; Brigham, Mark D.;
Trevino, Alexandro E.; Hsu, Patrick D.;
Heidenreich, Matthias; Cong, Le; Platt, Randall J.;
Scott, David A.; Church, George M. (2013-08-22).
"Optical control of mammalian endogenous
transcription and epigenetic states" . Nature.
500 (7463): 472–476.
doi:10.1038/nature12466 . ISSN 1476-4687 .
PMC 3856241  . PMID 23877069 .
95. Shimizu-Sato, Sae; Huq, Enamul; Tepperman,
James M.; Quail, Peter H. (2002-10-01). "A light-
switchable gene promoter system". Nature
Biotechnology. 20 (10): 1041–1044.
doi:10.1038/nbt734 . ISSN 1087-0156 .
PMID 12219076 .
96. Bugaj, Lukasz J.; Choksi, Atri T.; Mesuda,
Colin K.; Kane, Ravi S.; Schaffer, David V. (2013-
03-01). "Optogenetic protein clustering and
signaling activation in mammalian cells". Nature
Methods. 10 (3): 249–252.
doi:10.1038/nmeth.2360 . ISSN 1548-7105 .
PMID 23377377 .
97. Zhou, Xin X.; Chung, Hokyung K.; Lam, Amy
J.; Lin, Michael Z. (2012-11-09). "Optical control
of protein activity by fluorescent protein
domains" . Science. 338 (6108): 810–814.
doi:10.1126/science.1226854 . ISSN 1095-
9203 . PMC 3702057  . PMID 23139335 .
98. Wu, Yi I.; Frey, Daniel; Lungu, Oana I.; Jaehrig,
Angelika; Schlichting, Ilme; Kuhlman, Brian;
Hahn, Klaus M. (2009-09-03). "A genetically
encoded photoactivatable Rac controls the
motility of living cells" . Nature. 461 (7260):
104–108. doi:10.1038/nature08241 . ISSN 1476-
4687 . PMC 2766670  . PMID 19693014 .
99. Smart, Ashley D.; Pache, Roland A.; Thomsen,
Nathan D.; Kortemme, Tanja; Davis, Graeme W.;
Wells, James A. (2017-09-11). "Engineering a
light-activated caspase-3 for precise ablation of
neurons in vivo" . Proceedings of the National
Academy of Sciences of the United States of
America. 114 (39): E8174–E8183.
doi:10.1073/pnas.1705064114 . ISSN 1091-
6490 . PMC 5625904  . PMID 28893998 .
100. Purvis, Jeremy E.; Lahav, Galit (2013-02-28).
"Encoding and decoding cellular information
through signaling dynamics" . Cell. 152 (5): 945–
956. doi:10.1016/j.cell.2013.02.005 . ISSN 1097-
4172 . PMC 3707615  . PMID 23452846 .
101. Shimizu, Thomas S.; Tu, Yuhai; Berg,
Howard C. (2010-06-22). "A modular gradient-
sensing network for chemotaxis in Escherichia
coli revealed by responses to time-varying
stimuli" . Molecular Systems Biology. 6: 382.
doi:10.1038/msb.2010.37 . ISSN 1744-4292 .
PMC 2913400  . PMID 20571531 .
102. Santos, Silvia D. M.; Verveer, Peter J.;
Bastiaens, Philippe I. H. (2007-03-01). "Growth
factor-induced MAPK network topology shapes
Erk response determining PC-12 cell fate" .
Nature Cell Biology. 9 (3): 324–330.
doi:10.1038/ncb1543 . ISSN 1465-7392 .
103. Toettcher, Jared E.; Weiner, Orion D.; Lim,
Wendell A. (2013-12-05). "Using optogenetics to
interrogate the dynamic control of signal
transmission by the Ras/Erk module" . Cell. 155
(6): 1422–1434. doi:10.1016/j.cell.2013.11.004 .
ISSN 1097-4172 . PMC 3925772  .
PMID 24315106 .
Additional reading
Airan, R. D.; Hu, E. S.; Vijaykumar, R.; Roy, M.;
Meltzer, L. A.; Deisseroth, K. (October 2007).
"Integration of light-controlled neuronal firing
and fast circuit imaging" . Current Opinion in
Neurobiology. 17 (5): 587–92.
doi:10.1016/j.conb.2007.11.003 .
PMID 18093822 .
Alilain, W. J.; Li, X.; Horn, K. P.; Dhingra, R.; et
al. (November 2008). "Light-induced rescue
of breathing after spinal cord injury" . J.
Neurosci. 28 (46): 11862–70.
doi:10.1523/JNEUROSCI.3378-08.2008 .
PMC 2615537  . PMID 19005051 .
Arenkiel, B. R.; Peca, J.; Davison, I. G.;
Feliciano, Catia; et al. (April 2007). "In vivo
light-induced activation of neural circuitry in
transgenic mice expressing
channelrhodopsin-2" . Neuron. 54 (2): 205–
18. doi:10.1016/j.neuron.2007.03.005 .
PMID 17442243 .
Atasoy, D.; Aponte, Y.; Su, H. H.; Sternson, S.
M. (July 2008). "A FLEX switch targets
Channelrhodopsin-2 to multiple cell types for
imaging and long-range circuit mapping" . J.
Neurosci. 28 (28): 7025–30.
doi:10.1523/JNEUROSCI.1954-08.2008 .
PMC 2593125  . PMID 18614669 .
Ayling, O. G.; Harrison, T. C.; Boyd, J. D.;
Goroshkov, A.; Murphy, T. H. (March 2009).
"Automated light-based mapping of motor
cortex by photoactivation of
channelrhodopsin-2 transgenic mice". Nat.
Methods. 6 (3): 219–24.
doi:10.1038/nmeth.1303 . PMID 19219033 .
Berndt, A.; Yizhar, O.; Gunaydin, L. A.;
Hegemann, P.; Deisseroth, K. (February 2009).
"Bi-stable neural state switches" . Nature
Neuroscience. 12 (2): 229–34.
doi:10.1038/nn.2247 . PMID 19079251 .
Bi, A.; Cui, J.; Ma, Y. P.; Olshevskaya, Elena; et
al. (April 2006). "Ectopic expression of a
microbial-type rhodopsin restores visual
responses in mice with photoreceptor
degeneration" . Neuron. 50 (1): 23–33.
doi:10.1016/j.neuron.2006.02.026 .
PMC 1459045  . PMID 16600853 .
Busskamp, V.; Duebel, J.; Balya, D.; Fradot, M.;
Viney, T. J.; Siegert, S.; Groner, A. C.; Cabuy, E.;
Forster, V.; Seeliger, M.; Biel, M.; Humphries,
P.; Paques, M.; Mohand-Said, S.; Trono, D.;
Deisseroth, K.; Sahel, J. A.; Picaud, S.; Roska,
B. (2010-07-23). "Genetic reactivation of cone
photoreceptors restores visual responses in
retinitis pigmentosa" . Science. 329 (5990):
413–7. Bibcode:2010Sci...329..413B .
doi:10.1126/science.1190897 .
PMID 20576849 .
Cardin, J. A.; Carlén, M.; Meletis, K.; Knoblich,
U.; Zhang, F.; Deisseroth, K.; Tsai, L. H.; Moore,
C. I. (2010). "Targeted optogenetic
stimulation and recording of neurons in vivo
using cell-type-specific expression of
Channelrhodopsin-2" . Nature Protocols. 5
(2): 247–54. doi:10.1038/nprot.2009.228 .
PMID 20134425 .
Carter, M. E.; Adamantidis, A.; Ohtsu, H.;
Deisseroth, K.; de Lecea, L. (2009-09-02).
"Sleep homeostasis modulates hypocretin-
mediated sleep-to-wake transitions" . Journal
of Neuroscience. 29 (35): 10939–49.
doi:10.1523/JNEUROSCI.1205-09.2009 .
PMID 19726652 .
Carter, M. E.; Yizhar, O.; Chikahisa, S.; Nguyen,
H.; Adamantidis, A.; Nishino, S.; Deisseroth,
K.; de Lecea, L. (December 2010). "Tuning
arousal with optogenetic modulation of locus
coeruleus neurons" . Nature Neuroscience. 13
(12): 1526–33. doi:10.1038/nn.2682 .
PMC 3174240  . PMID 21037585 .
Chow, B. Y.; Han, X.; Dobry, A. S.; Qian,
Xiaofeng; et al. (January 2010). "High-
performance genetically targetable optical
neural silencing by light-driven proton
pumps" . Nature. 463 (7277): 98–102.
Bibcode:2010Natur.463...98C .
doi:10.1038/nature08652 . PMC 2939492  .
PMID 20054397 .
Claridge-Chang, A.; Roorda, R. D.; Vrontou, E.;
Sjulson, L.; Li, H.; Hirsh, J.; Miesenböck, G.
(October 2009). "Writing memories with light-
addressable reinforcement circuitry" . Cell.
139 (2): 405–15.
doi:10.1016/j.cell.2009.08.034 .
PMID 19837039 .
Clyne, J. D.; Miesenböck, G. (April 2008). "Sex-
specific control and tuning of the pattern
generator for courtship song in Drosophila".
Cell. 133 (2): 354–63.
doi:10.1016/j.cell.2008.01.050 .
PMID 18423205 .
Deisseroth, Karl. "Optogenetics: Controlling
the Brain with Light" .
Diester, I.; Kaufman, M. T.; Mogri, M.; Pashaie,
R.; Goo, W.; Yizhar, O.; Ramakrishnan, C.;
Deisseroth, K.; Shenoy, K. V. (March 2011).
"An optogenetic toolbox designed for
primates" . Nature Neuroscience. 14 (3): 387–
97. doi:10.1038/nn.2749 . PMC 3150193  .
PMID 21278729 .
Douglass, A. D.; Kraves, S.; Deisseroth, K.;
Schier, A. F.; Engert, F. (August 2008). "Escape
behavior elicited by single, channelrhodopsin-
2-evoked spikes in zebrafish somatosensory
neurons" . Curr. Biol. 18 (15): 1133–7.
doi:10.1016/j.cub.2008.06.077 .
PMC 2891506  . PMID 18682213 .
Gradinaru, V.; Zhang, F.; Ramakrishnan, C.;
Mattis, J.; Prakash, R.; Diester, I.; Goshen, I.;
Thompson, K. R.; Deisseroth, K. (2010-04-02).
"Molecular and cellular approaches for
diversifying and extending optogenetics" .
Cell. 141 (1): 154–65.
doi:10.1016/j.cell.2010.02.037 .
PMID 20303157 .
Gourine, A. V.; Kasymov, V.; Marina, N.; Tang,
F.; Figueiredo, M. F.; Lane, S.; Teschemacher,
A. G.; Spyer, K. M.; Deisseroth, K.; Kasparov, S.
(2010-07-30). "Astrocytes control breathing
through pH-dependent release of ATP" .
Science. 329 (5991): 571–5.
Bibcode:2010Sci...329..571G .
doi:10.1126/science.1190721 .
PMC 3160742  . PMID 20647426 .
Gunaydin, L. A.; Yizhar, O.; Berndt, A.; Sohal, V.
S.; Deisseroth, K.; Hegemann, P. (March
2010). "Ultrafast optogenetic control" . Nature
Neuroscience. 13 (3): 387–92.
doi:10.1038/nn.2495 . PMID 20081849 .
Han, X.; Boyden E. S.; Boyden (2007).
Rustichini, Aldo, ed. "Multiple-color optical
activation, silencing, and desynchronization
of neural activity, with single-spike temporal
resolution" . PLoS ONE. 2 (3): e299.
Bibcode:2007PLoSO...2..299H .
doi:10.1371/journal.pone.0000299 .
PMC 1808431  . PMID 17375185 .
Han, X.; Qian, X.; Bernstein, J. G.; Zhou, Hui-
hui; et al. (April 2009). "Millisecond-timescale
optical control of neural dynamics in the
nonhuman primate brain" . Neuron. 62 (2):
191–8. doi:10.1016/j.neuron.2009.03.011 .
PMC 2830644  . PMID 19409264 .
Hira, R.; Honkura, N.; Noguchi, J.; Maruyama,
Yoshio; et al. (May 2009). "Transcranial
optogenetic stimulation for functional
mapping of the motor cortex" . J. Neurosci.
Methods. 179 (2): 258–63.
doi:10.1016/j.jneumeth.2009.02.001 .
PMID 19428535 .
Hu, E. S.; Airan, R. D.; Vijaykumar, R.;
Deisseroth, K. (July 2008). "Brain circuit
dynamics" . The American Journal of
Psychiatry. 165 (7): 800.
doi:10.1176/appi.ajp.2008.08050764 .
PMID 18593784 .
Huber, D.; Petreanu, L.; Ghitani, N.; Ranade,
Sachin; et al. (January 2008). "Sparse optical
microstimulation in barrel cortex drives
learned behaviour in freely moving mice" .
Nature. 451 (7174): 61–4.
Bibcode:2008Natur.451...61H .
doi:10.1038/nature06445 . PMC 3425380  .
PMID 18094685 .
Hwang, R. Y.; Zhong, L.; Xu, Y.; Johnson, T.;
Zhang, F.; Deisseroth, K.; Tracey, W. D. (2007-
12-18). "Nociceptive neurons protect
Drosophila larvae from parasitoid wasps" .
Current Biology. 17 (24): 2105–16.
doi:10.1016/j.cub.2007.11.029 .
PMC 2225350  . PMID 18060782 .
Kuhlman, S. J.; Huang, Z. J.; Huang (2008).
Wong, Rachel O. L., ed. "High-resolution
labeling and functional manipulation of
specific neuron types in mouse brain by Cre-
activated viral gene expression" . PLoS ONE.
3 (4): e2005. Bibcode:2008PLoSO...3.2005K .
doi:10.1371/journal.pone.0002005 .
PMC 2289876  . PMID 18414675 .
Lagali, P. S.; Balya, D.; Awatramani, G. B.;
Münch, Thomas A; et al. (June 2008). "Light-
activated channels targeted to ON bipolar
cells restore visual function in retinal
degeneration". Nat. Neurosci. 11 (6): 667–75.
doi:10.1038/nn.2117 . PMID 18432197 .
Lee, J. H.; Durand, R.; Gradinaru, V.; Zhang, F.;
Goshen, I.; Kim, D. S.; Fenno, L. E.;
Ramakrishnan, C.; Deisseroth, K. (2010-06-
10). "Global and local fMRI signals driven by
neurons defined optogenetically by type and
wiring" . Nature. 465 (7299): 788–92.
Bibcode:2010Natur.465..788L .
doi:10.1038/nature09108 . PMC 3177305  .
PMID 20473285 .
Li, H. H.; Roy, M.; Kuscuoglu, U.; Spencer, C.
M.; Halm, B.; Harrison, K. C.; Bayle, J. H.;
Splendore, A.; Ding, F.; Meltzer, L. A.; Wright,
E.; Paylor, R.; Deisseroth, K.; Francke, U. (April
2009). "Induced chromosome deletions
cause hypersociability and other features of
Williams-Beuren syndrome in mice" . EMBO
Molecular Medicine. 1 (1): 50–65.
doi:10.1002/emmm.200900003 .
PMC 3378107  . PMID 20049703 .
Liewald, J. F.; Brauner, M.; Stephens, G. J.;
Bouhours, Magali; et al. (October 2008).
"Optogenetic analysis of synaptic function".
Nat. Methods. 5 (10): 895–902.
doi:10.1038/nmeth.1252 . PMID 18794862 .
Lima, S. Q.; Hromádka, T.; Znamenskiy, P.;
Zador, A. M.; Hromádka; Znamenskiy; Zador
(2009). Nitabach, Michael N., ed. "PINP: a
new method of tagging neuronal populations
for identification during in vivo
electrophysiological recording" . PLoS ONE. 4
(7): e6099. Bibcode:2009PLoSO...4.6099L .
doi:10.1371/journal.pone.0006099 .
PMC 2702752  . PMID 19584920 .
Lin, J. Y.; Lin, M. Z.; Steinbach, P.; Tsien, R. Y.;
Lin; Steinbach; Tsien (March 2009).
"Characterization of engineered
channelrhodopsin variants with improved
properties and kinetics" . Biophys. J. 96 (5):
1803–14. Bibcode:2009BpJ....96.1803L .
doi:10.1016/j.bpj.2008.11.034 .
PMC 2717302  . PMID 19254539 .
Liu, Q.; Hollopeter, G.; Jorgensen, E. M.;
Hollopeter; Jorgensen (June 2009). "Graded
synaptic transmission at the Caenorhabditis
elegans neuromuscular junction" . Proc. Natl.
Acad. Sci. U.S.A. 106 (26): 10823–8.
Bibcode:2009PNAS..10610823L .
doi:10.1073/pnas.0903570106 .
PMC 2705609  . PMID 19528650 .
Llewellyn, M. E.; Thompson, K. R.; Deisseroth,
K.; Delp, S. L. (October 2010). "Orderly
recruitment of motor units under optical
control in vivo" . Nature Medicine. 16 (10):
1161–5. doi:10.1038/nm.2228 .
PMID 20871612 .
Lobo, M. K.; Covington, H. E., 3rd; Chaudhury,
D.; Friedman, A. K.; Sun, H.; Damez-Werno, D.;
Dietz, D. M.; Zaman, S.; Koo, J. W.; Kennedy, P.
J.; Mouzon, E.; Mogri, M.; Neve, R. L.;
Deisseroth, K.; Han, M. H.; Nestler, E. J. (2010-
10-15). "Cell type-specific loss of BDNF
signaling mimics optogenetic control of
cocaine reward" . Science. 330 (6002): 385–
90. Bibcode:2010Sci...330..385L .
doi:10.1126/science.1188472 .
PMC 3011229  . PMID 20947769 .
Miesenböck, G. (October 2008). "Lighting up
the brain". Sci. Am. 299 (4): 52–9.
doi:10.1038/scientificamerican1008-52 .
PMID 18847085 .
Miesenböck, G. (October 2009). "The
optogenetic catechism". Science. 326 (5951):
395–9. Bibcode:2009Sci...326..395M .
doi:10.1126/science.1174520 .
PMID 19833960 .
Miller, G. (December 2006). "Optogenetics.
Shining new light on neural circuits" .
Science. 314 (5806): 1674–6.
doi:10.1126/science.314.5806.1674 .
PMID 17170269 .
Schneider, M. B.; Gradinaru, V.; Zhang, F.;
Deisseroth, K. (May 2008). "Controlling
neuronal activity" . The American Journal of
Psychiatry. 165 (5): 562.
doi:10.1176/appi.ajp.2008.08030444 .
PMID 18450936 .
Schröder-Lang, S.; Schwärzel, M.; Seifert, R.;
Strünker, Timo; et al. (January 2007). "Fast
manipulation of cellular cAMP level by light in
vivo". Nature Methods. 4 (1): 39–42.
doi:10.1038/nmeth975 . PMID 17128267 .
Szobota, S.; Gorostiza, P.; Del Bene, F.; Wyart,
Claire; et al. (May 2007). "Remote control of
neuronal activity with a light-gated glutamate
receptor" . Neuron. 54 (4): 535–45.
doi:10.1016/j.neuron.2007.05.010 .
PMID 17521567 .
Toni, N.; Laplagne, D. A.; Zhao, C.; Lombardi,
Gabriela; et al. (August 2008). "Neurons born
in the adult dentate gyrus form functional
synapses with target cells" . Nat. Neurosci.
11 (8): 901–7. doi:10.1038/nn.2156 .
PMC 2572641  . PMID 18622400 .
Tønnesen, J.; Sørensen, A. T.; Deisseroth, K.;
Lundberg, C.; Kokaia, M. (2009-07-21).
"Optogenetic control of epileptiform activity" .
Proceedings of the National Academy of
Sciences of the United States of America. 106
(29): 12162–7.
Bibcode:2009PNAS..10612162T .
doi:10.1073/pnas.0901915106 .
PMC 2715517  . PMID 19581573 .
Wang, S.; Szobota, S.; Wang, Y.; Volgraf,
Matthew; et al. (December 2007). "All optical
interface for parallel, remote, and
spatiotemporal control of neuronal activity".
Nano Lett. 7 (12): 3859–63.
Bibcode:2007NanoL...7.3859W .
doi:10.1021/nl072783t . PMID 18034506 .
Wang, H.; Peca, J.; Matsuzaki, M.; Matsuzaki,
K.; et al. (May 2007). "High-speed mapping of
synaptic connectivity using photostimulation
in Channelrhodopsin-2 transgenic mice" .
Proc. Natl. Acad. Sci. U.S.A. 104 (19): 8143–8.
Bibcode:2007PNAS..104.8143W .
doi:10.1073/pnas.0700384104 .
PMC 1876585  . PMID 17483470 .
Wang, Y.; Dye, C. A.; Sohal, V.; Long, J. E.;
Estrada, R. C.; Roztocil, T.; Lufkin, T.;
Deisseroth, K.; Baraban, S. C.; Rubenstein, J.
L. (2010-04-14). "Dlx5 and Dlx6 regulate the
development of parvalbumin-expressing
cortical interneurons" . Journal of
Neuroscience. 30 (15): 5334–45.
doi:10.1523/JNEUROSCI.5963-09.2010 .
PMC 2919857  . PMID 20392955 .
Weick, J. P.; Johnson, M. A.; Skroch, S. P.;
Williams, J. C.; Deisseroth, K.; Zhang, S. C.
(November 2010). "Functional control of
transplantable human ESC-derived neurons
via optogenetic targeting" . Stem cells
(Dayton, Ohio). 28 (11): 2008–16.
doi:10.1002/stem.514 . PMC 2988875  .
PMID 20827747 .
Zhang, F.; Wang, L. P.; Boyden, E. S.;
Deisseroth, K. (October 2006).
"Channelrhodopsin-2 and optical control of
excitable cells". Nat. Methods. 3 (10): 785–
92. doi:10.1038/nmeth936 .
PMID 16990810 .
Zhang, F.; Wang, L. P.; Brauner, M.; Liewald,
Jana F.; et al. (April 2007). "Multimodal fast
optical interrogation of neural circuitry".
Nature. 446 (7136): 633–9.
Bibcode:2007Natur.446..633Z .
doi:10.1038/nature05744 . PMID 17410168 .
Zhang, F.; Aravanis, A. M.; Adamantidis, A.; de
Lecea, L.; Deisseroth, K. (August 2007).
"Circuit-breakers: optical technologies for
probing neural signals and systems". Nature
Reviews Neuroscience. 8 (8): 577–81.
doi:10.1038/nrn2192 . PMID 17643087 .
Zhang, Y. P.; Holbro, N.; Oertner, T. G.; Holbro;
Oertner (August 2008). "Optical induction of
plasticity at single synapses reveals input-
specific accumulation of alphaCaMKII" .
Proc. Natl. Acad. Sci. U.S.A. 105 (33): 12039–
44. Bibcode:2008PNAS..10512039Z .
doi:10.1073/pnas.0802940105 .
PMC 2575337  . PMID 18697934 .
Zhang, F.; Prigge, M.; Beyrière, F.; Tsunoda,
Satoshi P; et al. (June 2008). "Red-shifted
optogenetic excitation: a tool for fast neural
control derived from Volvox carteri" . Nat.
Neurosci. 11 (6): 631–3.
doi:10.1038/nn.2120 . PMC 2692303  .
PMID 18432196 .
Zhang, F.; Gradinaru, V.; Adamantidis, A. R.;
Durand, R.; Airan, R. D.; de Lecea, L.;
Deisseroth, K. (2010). "Optogenetic
interrogation of neural circuits: technology for
probing mammalian brain structures" . Nature
Protocols. 5 (3): 439–56.
doi:10.1038/nprot.2009.226 .
PMID 20203662 .
Zhang, J.; Laiwalla, F.; Kim, J. A.; Urabe, H.;
Van Wagenen, R.; Song, Y. K.; Connors, B. W.;
Zhang, F.; Deisseroth, K.; Nurmikko, A. V.
(October 2009). "Integrated device for optical
stimulation and spatiotemporal electrical
recording of neural activity in light-sensitized
brain tissue" . Journal of neural engineering. 6
(5): 055007. Bibcode:2009JNEng...6e5007Z .
doi:10.1088/1741-2560/6/5/055007 .
PMC 2921864  . PMID 19721185 .
Zhu, P.; Narita, Y.; Bundschuh, S. T.; Fajardo,
O.; Schärer, Y. P.; Chattopadhyaya, B.;
Bouldoires, E. A.; Stepien, A. E.; Deisseroth, K.;
Arber, S.; Sprengel, R.; Rijli, F. M.; Friedrich, R.
W. (2009-12-11). "Optogenetic Dissection of
Neuronal Circuits in Zebrafish using Viral
Gene Transfer and the Tet System" . Frontiers
in Neural Circuits. 3: 21.
doi:10.3389/neuro.04.021.2009 .
PMC 2805431  . PMID 20126518 .
Zimmermann, G.; Wang, L. P.; Vaughan, A. G.;
Manoli, D. S.; Zhang, F.; Deisseroth, K.; Baker,
B. S.; Scott, M. P. (2009). Nitabach, Michael
N., ed. "Manipulation of an innate escape
response in Drosophila: photoexcitation of
acj6 neurons induces the escape response" .
PLoS ONE. 4 (4): e5100.
Bibcode:2009PLoSO...4.5100Z .
doi:10.1371/journal.pone.0005100 .
PMC 2660433  . PMID 19340304 .

External links
Optogenetics Resource Center , maintained
by the Deisseroth lab.
Synthetic Neurobiology Group, MIT , the
portal of the Boyden lab.
OpenOptogenetics.org , an optogenetics wiki,
and its companion blog .
Molecular Neurogenetics and Optophysiology
Laboratory ,"Optogenetic activation and
silencing recordings of individual prefrontal
cortical neurons in vivo and in vitro.
Sohal lab portal
Nurmikko lab portal
Lab of Dr. Zhuo-Hua Pan
Optophysiology at the Tyler lab
Video: Ed Boyden on Optogenetics -- selective
brain stimulation with light (SPIE Newsroom,
April 2011)

Retrieved from "https://en.wikipedia.org/w/index.php?


title=Optogenetics&oldid=823762315"

Last edited 16 days ago by FrescoBot

Content is available under CC BY-SA 3.0 unless


otherwise noted.

Vous aimerez peut-être aussi