You are on page 1of 16

REVIEWS

NEURAL CIRCUITS

Optogenetic investigation of neural


circuits underlying brain disease in
animal models
Kay M.Tye1,2 and Karl Deisseroth1,3,4,5
Abstract | Optogenetic tools have provided a new way to establish causal relationships
between brain activity and behaviour in health and disease. Although no animal model
captures human disease precisely, behaviours that recapitulate disease symptoms may be
elicited and modulated by optogenetic methods, including behaviours that are relevant to
anxiety, fear, depression, addiction, autism and parkinsonism. The rapid proliferation of
optogenetic reagents together with the swift advancement of strategies for implementation
has created new opportunities for causal and precise dissection of the circuits underlying
brain diseases in animal models.

To improve understanding of psychiatric and neurologi- electrophysiological, pharmacological and behavioural


Opsins
Membrane-bound proteins cal disorders, it will be important to identify the under- assessments. We also highlight the advantages and
that can incorporate small lying neural circuits, to pinpoint the precise nature of practical limitations of these approaches for the study
organic retinal molecules to the causally important aberrations in these circuits of psychiatric and neurological disease.
become a light receptor. and to modulate circuit and behavioural dysfunction
with precise and specific experimental interventions. Technological advances in optogenetics
However, such a deep, circuit-level understanding of The optogenetic toolbox includes a rapidly expand-
neuropsychiatric disorders, or indeed even of normal ing array of available opsin variants that offer both
CNS circuit function, has been challenging to achieve distinct advantages and individual limitations in
with traditional methods. The complexity of neural cir- controlling cellular activity or signalling 3,1221. Other
1
Department of
Bioengineering, Stanford
cuitry has historically precluded the use of genetically important components of the toolbox are light-delivery
University, 318 Campus Drive, and temporally precise manipulations to probe detailed methods6,9,2228, targeting strategies16,2931 and trans-
Clark Center, Stanford, mechanisms of function and dysfunction. genic rodent lines that increase the range of available
California 94305-5444, USA. Optogenetics1,2 describes the now widespread use specific cellular targets3234. For example, the recent
2
Picower Institute of Learning
of microbial opsins3, or related tools4, that can be acti- development of devices35,36 and transgenic rat lines37
and Memory, Department of
Brain and Cognitive Sciences, vated by illumination to manipulate cells with high that facilitate integration of optogenetic techniques
Massachusetts Institute of specificity and temporal precision57 even within intact with measures of neural activity have advanced the
Technology, Cambridge, tissue or behaving animals811. Here, we briefly review application of optogenetic tools to investigate the neu-
Massachusetts how optogenetic approaches have been used to dissect ral bases of complex behaviours that are relevant to
02139-4307, USA.
3
Department of Psychiatry,
neural circuits in animal models of symptoms that are neuropsychiatricdisease.
Stanford University, 401 relevant to fear, anxiety, depression, schizophrenia,
Quarry Road, Stanford, addiction, social dysfunction, Parkinsons disease and Integration of optogenetics with mapping techniques.
California 94305-5717, USA. epilepsy. Successful probing of complexdiseases in The recent integration of fMRI with optogenetic manip-
4
CNC Program, Stanford
this way will depend on the validity of animal mod- ulation, now referred to as ofMRI, has not only vali-
University, Stanford,
California 94305, USA. els used to identify the crucial circuit elements and dated a previously assumed interpretation of the fMRI
5
Howard Hughes Medical activity patterns that are involved in each cluster of BOLD signal38 (that increased neuronal activity in local
Institute, Stanford University symptoms, and the precision and efficiency of inter- excitatory neurons can causally trigger, rather than
Medical School, Stanford, ventions designed to selectively target these elements simply correlate with, an increase in the local BOLD
California 94305-5323, USA.
emails: kaytye@mit.edu;
or patterns. Therefore, we also discuss new strategies signal) but has also shown that it is possible to assay
deissero@stanford.edu for targeting opsins to specific cells or circuit ele- the effects of precise optogenetic manipulations on
doi:10.1038/nrn3171 ments and principles for integrating optogenetics with global brain activity. Given that many neuropsychiatric

NATURE REVIEWS | NEUROSCIENCE VOLUME 13 | APRIL 2012 | 251

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

diseases are likely to involve distributed perturbations, As SSFO also has an enhanced sensitivity to light, it
global approaches such as ofMRI may be crucial for enables the non-invasive light-induced activation of
identifying and mapping the downstream effects of SSFO-expressing neurons up to 3mm below the sur-
cell-type or projection-specific manipulations (in an face of the brain when an optical fibre is placed just
unbiased fashion). above the brain surface48. Thus, the development of the
Local, detailed circuit-mapping has also benefited SSFO may facilitate research in large brain regions or
greatly from optogenetics. Continuing a long-standing in large-brainedanimals.
tradition of mapping neural circuitry in mammals Another group of noteworthy opsins is the red-
with optical approaches39, and in certain cases using shifted activation wavelength ChR1/VChR1 chimaera
new classes of light delivery 40, several elegant optoge- (C1V1) family, which includes variants that are sig-
netic studies have already made substantial advances nificantly more potent48 than ChR2 and approximately
in detailed circuit mapping 4143. These studies have fourfold more potent than the Volvox channelrho-
helped to clarify the role of specific cortical layers in dopsin 1 (VChR1), a red-shifted opsin developed
the regulation of activity flow, as well as to delineate previously 14. Members of the C1V1 family and their
the detailed pattern of synaptic inputs arising from associated variants have a peak activation wavelength
distinct cortical layers onto distinct subcellular loca- of ~560nm and can readily be activated by 590nm
tions in neocortical principal cells. By providing a rich light, thus increasing the feasibility of combinatorial
fMRI
source of information that would have been difficult excitation or depolarization of different populations of
(Functional magnetic
resonance imaging). This or impossible to obtain by other means, these studies neurons at distinct experimental epochs or patterns21,48.
method can use detection of may lay the groundwork for identifying circuit or con- These opsins have been used to investigate behaviours
blood oxygen levels as a proxy nectivity phenotypes that can go awry in diseasestates. that are relevant to autism and schizophrenia48, and are
for neural activity, and offers a promising candidates for the study of other diseases of
non-invasive method to
globally assay brain activity in
New opsin variants. Earlier optogenetic tools, such thebrain.
humans. as channelrhodopsin 2 (ChR2) 5,13 which enables
action potential elicitation to be time-locked to light Use of transgenic rodents in optogenetics. Tools for
BOLD pulses or halorhodopsin (NpHR)16,18,4446 and pro- conferring genetic specificity to opsin delivery are also
(Blood oxygen level
ton pumps 16,19,21 which enable hyperpolarization improving. A large number of Cre recombinase-driver
dependent). The BOLD signal
is one kind of signal that fMRI of membranes to inhibit the production of action mouse lines have already been used in optogenetic
can use to assess neural potentials are still useful. However, the expan- research (reviewed in REF. 6). Although transgenic
activity. sion of the optogenetic toolbox (FIG.1) now provides mouse lines have proven to be very useful in the study
greater flexibility in experimental design and more of cells, circuits and behaviours that are relevant to
Channelrhodopsin
A light-driven cation channel,
powerful and refined manipulations. For example, disease31,4855, the more complex behavioural and elec-
found in algae, that can be engineered channelrhodopsin variants (including the trophysiological assays available in rats could provide
used to depolarize cell ChETA family 20,21 and ChIEF47) can be used to evoke important additional insight. However, optogenetic
membranes. ultra-fast firing frequencies (up to 200Hz or more) in research in rats has been hampered by a lack of genetic
fast-spikingneurons. tools for targeting opsins to specific cell types that are
Halorhodopsin
A light-driven chloride ion Although the ability to elicit action potentials that implicated in disease. The recent development of two
pump found in phylogenetically are time-locked to light pulses is powerful, the syn- Cre recombinase-driver rat lines targeting tyrosine
ancient archaea, known as chrony and patterning of an experimentally delivered hydroxylase and choline acetyltransferase neurons,
halobacteria, that can be used illumination pattern may not represent the physiologi- with cell-type-specific promoter or enhancer regions
to hyperpolarize cell
membranes.
cal neural code. OptoXRs4 (opsinreceptor chimae- that were too large to package into most vectors37, has
ras in which the intracellular loops of rhodopsin are addressed this need and may facilitate research into
UP states replaced with intracellular loops from other G protein- animal models of psychiatric and neurological disease
Sub-threshold membrane coupled receptors such as adrenergic receptors) now states. These, and other new transgenic animals, will
depolarization states that have
allow light-activated initiation of specific G protein- set the stage for integrated studies combining invivo
been observed to
spontaneously occur invivo in coupled signalling cascades in targeted neurons within electrophysiological and optogenetic studies during
some neurons and that may freely moving mammals. This can lead to altered excit- sophisticated behavioural assays.
serve to increase the intrinsic ability in a population of cells without dictating precise
excitability of the neuron. neural spike times. Alternatively, to increase excitability Optogenetics in behavioural studies
Cre recombinase
of neurons without dictating a specific pattern of firing, Optogenetic approaches have already begun to alter
DNA recombinase that it is possible to apply a long-lasting and subthreshold the way in which traditional behavioural assays are
excises DNA sequences membrane depolarization, as seen in cortical UP states. used. In addition to cell-type-specific and projection-
flanked by loxP sequences with The step-function opsin (SFO)17 facilitates this kind specific targeting strategies that allow unprecedented
the same orientation, or inverts
of intervention by delivering a prolonged, bi-stable, precision in manipulation, the temporal properties of
sequences flanked by loxP
sites with opposite orientation. subthreshold depolarization of membranes. optogenetics are reshaping the style of experimental
It is effective in mammalian New opsins such as the stabilized step-function design. For example, the immediate reversibility of
cells invitro and invivo. opsin (SSFO) 48, which is a double mutant of ChR2 optogenetic manipulations, compared to traditional
(Asp156Ala and Cys128Ser), are a substantial improve- pharmacological techniques that require a lengthy
Vectors
Vehicles used to transfer
ment on the previous17 single mutant SFO (for example, wash-out period, allows multi-day tests to be con-
genetic material to a target Cys128Ser) in that the stability of the step function-like densed into a single session. The elevated plus
cell. depolarization is greater on the order of 30min17,21,48. maze with optogenetics now allows within-subject

252 | APRIL 2012 | VOLUME 13 www.nature.com/reviews/neuro

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

comparison of conditions (light on and light off) in a and a test day. As the traditional paradigm typically
single behavioural session56. Similarly, paradigms such involved the animal being locked into one compart-
as conditioned place preference (CPP) were traditionally ment of the chamber while receiving the treatment
performed with a habituation day, a conditioning day (such as a drug infusion) and then restricted in the

ChR NpHR BR/PR OptoXR

Gq Gs Gi

CI Na+ Ca2+ H+ K+ [InsP3] [cAMP] [cAMP]


[DAG]

Bacterial ATP
cyclase ATP
cAMP

b 650

Hyperpolarizing Blue depolarizing


Biochemical modulation Bi-stable depolarizing
Fast inhibition Red-shifted depolarizing ChETA variants
600 NpHR

C1V1
Arch
Peak activation (nm)

eBR E122T/E162T
Step function opsin (bi-stable depolarization)
550 VChR1

C1V1 VChR1 VChR1


C123S C123S/D151A
ChRGR* C1V1
L132C* E162T
500 CatCH Opto-2 ChR2
E123T Opto-2
ChIEF T159C Rh-CT ChR2 D156A/
C128S C128S
E123A ChR2 BlaC
ChR2 ChR2
450 E123T/ WT C128T bPAC C128A D156A
H134R
T159C
Biochemical modulation
Fast excitation

400
1 ms 10 ms 100 ms 1s 10 s 100 s 1,000 s 10,000 s
1 min 30 min
o
Figure 1 | Optogenetic tools. a | Major classes of single-component optogenetic tools include cation-permeable channels
for membrane depolarization (such as channelrhodopsins (ChRs)), chloride pumps (for example, halorhodopsin (NpHR)) and
Nature Reviews | Neuroscience
proton pumps (such as bacteriorhodopsin or proteorhodopsin (BR/PR)) for membrane hyperpolarization, and
light-activated membrane-bound G protein-coupled (OptoXR) or soluble (bacterial cyclase) receptors that mimic various
signalling cascades. b | Tools that have been characterized in terms of wavelength activation spectra and decay kinetics.
The chart shows peak activation wavelength plotted against decay kinetics and illustrates groupings of tools over the range
of spectral and temporal characteristics. This also demonstrates why it is feasible to use tools that are well separated in
Conditioned place spectral and/or temporal domains to achieve dual-channel control. It should be noted that the kinetics for OptoXR were
preference characterized invivo using an assay that measured a downstream readout (spiking) and probably represent an upper bound
(CPP). A behavioural test in for these properties. Decay kinetics are temperature-dependent; all reported values except for channelrhodopsin-green
which an unconditioned receiver (ChRGR) were recorded at room temperature (an ~50% decrease in decay kinetics is expected if the temperature is
stimulus is paired with one increased to 37C). ChRGR has only been studied at 34C. We have therefore extrapolated the likely range for this protein
distinctive context and a at room temperature. The decay kinetics for the L132C mutation (calcium translocating channelrhodopsin (CatCH)) were
neutral event is paired with a
not measured in neurons, and these properties may depend on factors including the presence of other channels in the host
different context. Preference
cell and the host cell tolerance of, and response to, elevated intracellular Ca2+ levels197. The recently determined198 crystal
is determined by allowing the
animal to move between
structure of ChR2 may allow the design of additional classes of optogenetic tools. The / indicates the combination of two
the two contexts and mutations. Figure is modified, with permission, from REF.6 (2011) Elsevier. Arch, archaerhodopsin; bPAC, bacterial
measuring the amount of time photoactivated adenylyl cyclase; C1V1, ChR1/VChR1 chimaera; cAMP, cyclic AMP; DAG, diacylglycerol; eBR, enhanced
spent in each context. bacteriorhodopsin; InsP3, inositol trisphosphate; VChR1, Volvox channelrhodopsin 1.

NATURE REVIEWS | NEUROSCIENCE VOLUME 13 | APRIL 2012 | 253

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Designing optogenetic experiments to study brain disease


or duty cycles, the light that is emitted from the optical
fibre may cause heating. Heating neurons may not only
There are at least five major steps in the design alter their activity in a nonspecific manner but may
of optogenetic experiments to study Select opsin
also be detrimental to cell health. Appropriate con-
behaviour in normal function or disease trols, as well as assessment of light source stability and
models (see the figure).
performance, must be carefully and frequently exam-
Select the opsin best suited to the Target cell type ined to ensure precise and reliable light output and
experimental goals. There are trade-offs for interpretation of light effects6.
different aspects of performance, such as peak
Another limitation of optogenetic tools is the poten-
photocurrent, kinetics, activation wavelength
spectrum and light sensitivity.
tial for toxicity at very high expression levels or long-
Delivery of light
term expression. As expression of a microbial opsin
Select targeting strategy or vector to express
typically means the insertion of light-activated chan-
opsin in target cells. Many kinds of viral
nels or pumps into the cell membrane, there may be a
transduction can restrict cell-body expression
of opsin to the injection site, and can be used Set temporal parameters maximal level of opsin expression that can be tolerated
in wild-type animals or Cre recombinase lines. by a given cell. To determine whether opsin expres-
Some transgenic lines constitutively express sion has altered cell health, it is necessary to perform
opsin. controls under the same conditions as the experimen-
Plan validation tal parameters. Cell health and all other performance
Select light delivery method. Fibre optics are
the most common method of light delivery to parameters will vary with many parameters, including
deep cell bodies or axon terminals. The numerical aperture, diameter andReviews
Nature mode of| the light intensity, virus titre, injection volume, vector,
Neuroscience
fibre will influence the spread of light. There are trade-offs for using acute or chronically opsin, cell type, species and incubation time21. Finally,
implantable fibres, as acute fibres allow for pharmacological manipulations but are transient intracellular or extracellular ion balance
delicate and easy to break. changes (over seconds) may occur after modulated (or
Choose appropriate temporal parameters. Duty cycle, pulse duration, frequency and natural) activity patterns, and optogenetic experiments
epoch pattern are the key light-delivery parameters to select. Depending on the typically are designed with this in mind. It is important
behavioural assay, exploring within-session light manipulations can maximize the utility to consider the contribution of many parameters dur-
of optogenetic tools. ing the growth of this new field, and we recommend
Validate the experimental manipulation. To verify that the opsin, targeting strategy that experimenters empirically examine measures of
and illumination parameters are manipulating cells in the intended manner, interest in each new preparation.
confirmation using electrophysiology, immunohistochemistry or other measures is
crucial for data interpretation. Invivo optogenetic design
Designing invivo optogenetic experiments for inves-
tigation of the neural circuits underlying behaviour in
other compartment during the control treatment (such both health and disease requires a unique set of consid-
as saline infusion), the only behavioural readout of erations (BOX1). In selecting an appropriate opsin gene
the effect was on the test day. However, with the tem- for experiments, it is necessary to recognize the trade-
poral agility of optogenetic techniques, the condi- offs that accompany each choice. For example, there is
tioning day can now be performed with the animal an inverse correlation between light sensitivity and off-
moving freely and with light delivery only when the kinetics21; volume of activation could be sacrificed in
animal enters the conditioning chamber 4. This ena- favour of greater precision of temporal control or vice
bles the experimenter to quantify a location preference versa. Thus, it is important to consider which proper-
for the animal during conditioning, thus enriching ties are most important for each experimentalaim.
the data set by providing information about the time It is also important to determine the best targeting
course over which a conditioned preference devel- strategy (FIG.2). One commonly used strategy is viral
ops. Furthermore, animals can be allowed to freely transduction, an approach that will initially limit opsin
explore a chamber, and different light stimulation expression to the injection site. However, after some
parameters (duty cycle, pulse frequency, pulse dura- incubation time, viral transduction strategies enable
tion and intensity) can be triggered depending on the anterograde control capabilities6,7, meaning that the
animals location. This allows for a much finer measure protein is expressed in local cell bodies and trafficked
of optimal stimulation parameters in comparison to to the downstream terminals of those cell bodies; these
different drug doses, which cannot be tested within a projections can then be illuminated to control cells by
single session. This utility of the temporal resolution of virtue of their outgoing (efferent) connectivity. There
optogenetics is particularly transformative in experi- are many promoters available for transducing viruses
ments that use temporally specific variables, such as into wild-type animals6, and the most commonly used
discrete cues or tasks that have behaviours with critical viral vectors are lentivirus and adeno-associated viruses.
time windows, such as a decision point in a Tmaze. Other viruses that have potential utility include herpes
Duty cycle However, as well as the new advantages that opto simplex viruses (HSVs) or rabies viruses, which can
The time that a machine, genetic techniques offer, we are presented with several infect axon terminals and present retrograde-like tar-
system or light source spends
in an active state as a fraction
new limitations, caveats and considerations. One very geting opportunities for controlling afferents to a struc-
of the total time under important limitation to consider is the production of ture; however, there may be more toxicity associated
consideration. heat with illumination. When using high light powers with theseviruses.

254 | APRIL 2012 | VOLUME 13 www.nature.com/reviews/neuro

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

For promoter sequences that are too large to package optogenetic targeting in rodent anxiety models, a spe-
into a viral vector, Cre recombinase-driver lines used cific population of amygdala synapses has been identi-
in conjunction with Cre-dependent opsin-expressing fied that can rapidly and reversibly modulate baseline
viral vectors offer an attractive alternative, as first anxiety levels in a freely moving mammal56. This study
demonstrated behaviourally in tyrosine hydroxylase capitalized on the fact that microbial opsins can be
(TH)::Cre mice31 and parvalbumin (PV)::Cre mice53,54. expressed throughout the cell, including on the mem-
Many such targeting strategies exist 29,33,57, and there are branes of axons and axon terminals (a property that
also transgenic mice that constitutively express ChR2 may be enhanced by the inclusion of specific target-
in certain types of cells32,33,58. Opsin expression in these ing sequences16), allowing axon depolarization to be
mice is not subject to the variability that is associated generated simply by illumination of the axons them-
with viral infection or the burden of a lengthy incu- selves. The study also used a bevelled light-delivery
bation time. These advantages may be particularly cannula to guide light selectively to axons projecting
important for some applications; however, they are from one brain region to another. It was shown that
accompanied by the caveat that the illuminated region selective optogenetic stimulation (with a channelrho-
may contain not only local photosensitive targeted dopsin) of excitatory basolateral amygdala (BLA) cells
cell bodies but also photosensitive projections from cell with axons that project to the central nucleus of the
bodies that are located elsewhere in the brain. Another amygdala (CeA) produced an anxiolytic effect. This
recently developed strategy involves selectively infect- phenotype was markedly different from the anxiogenic
ing specific neuron types by using a viral vector that effect that was observed when BLA excitatory cell bod-
recognizes receptors on the exterior of the cell59,60. ies were illuminated nonspecifically without regard to
Lastly, it is crucial to determine the most appropri- projection target 56,69 (FIG.3). Indeed, BLA projection
ate method for delivery of light into the brain. Optical neurons have many targets, including not only the
fibres can be used either acutely (for example, a bare centrolateral and centromedial nuclei of the amygdala
fibre that is itself a patch cable can be inserted using but the nucleus accumbens (NAc), the prefrontal cortex,
a guide cannula) 9,10,25 or chronically (for example, the bed nucleus of the stria terminalis and many other
an implantable optical fibre can be joined to a patch structures that could have fundamentally different (and
cable outside the brain) 6,28. Acute optical fibres can even opposite) effects on anxiety. Thus, optogenetic
deliver pharmacological agents to the same location projection targeting enabled resolution of this distinct
as light 56,61; however, because of the delicate nature of endogenous pathway for anxiolysis in the amygdala.
optical fibres, the risk of fibres breaking in the guide Moreover, selective illumination of these BLACeA
cannula is a substantial disadvantage. Although chroni- axons expressing an enhanced version of the hyper-
cally implantable fibres do not allow for integration polarizing halorhodopsin, eNpHR3.0 (REF.16), induced
with pharmacology at the same site, they do offer an anxiogenic effect, establishing that this cellular
increased durability for multiple-day experiments6,28. projection is capable of bidirectional modulation of
Laser diodes are a widely used light source and are behaviours that are relevant to anxiety 56.
easy to couple to fibre optics6: light-emitting diodes Panic attacks are briefer and more intense than anxi-
(LEDs) can be used directly at the tissue (although ety episodes and are characterized by intense bursts
there are associated problems owing to high local heat of terror, apprehension and autonomic arousal, often
generation) or coupled to fibres in the same way as with chronic consequences such as a debilitating fear of
laser diodes (but with some light loss)22,26,62,63. Finally, future attacks64,65,67. Interestingly, these can be triggered
once an opsin is expressed and light is delivered to by isolated components (which are not threatening
the desired location, the light stimulation parameters in isolation) of a context that was previously associ-
should be considered. Possible issues include the effects ated with fear. Although it has long been thought that
of heating, light scattering and the physiological capac- the lateral amygdala is a crucial region for processing
ity of the targeted neurons. With so many variables, fear 7074, it had not been demonstrated directly that
it is important to carefully validate with imaging, activation of glutamatergic lateral amygdala neurons
physiology, or c-FOS staining that neurons are being alone could induce a fear response. Optogenetic tools
manipulated with the strength and specificity intended have now allowed researchers to selectively target the
before interpreting any experimentalresults. glutamatergic pyramidal neurons of the lateral amyg-
dala, which also contains local GABAergic interneu-
Circuitry of fear and anxiety disorders rons. Fear responses in mice were observed in response
Anxiety disorders, which include generalized anxiety to light-induced activation of ChR2expressing lateral
disorder, panic disorder, post-traumatic stress disorder amygdala neurons; moreover, pairing the presentation
(PTSD) and phobias, are the most common class of of a neutral stimulus (a tone) with lateral amygdala
Conditioned fear responses
A fear-associated stimulus psychiatric diseases, with a lifetime prevalence of ~28% illumination led to the observation of conditioned fear
(such as a shock-predictive (REFS 64,65). Although anxiety disorders are common, responses to the tonealone69.
tone) that may evoke the available treatments are inadequate in terms of Fearful responses to specific stimuli or contexts can
conditioned responses such as efficacy and side effects6668. also include phobias, which can arise from exposure to
freezing, fear-potentiated
startle or increases in blood
Anxiety is characterized by a sustained state of harm or threat and represent the single largest sub-class
pressure, perspiration or heart apprehension in the absence of an immediate threat 65. of anxiety disorders, affecting up to 12% of the popu-
rate. By establishing methods for projection-specific lation67. Although many lesion and pharmacological

NATURE REVIEWS | NEUROSCIENCE VOLUME 13 | APRIL 2012 | 255

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

Injection site Viral expression Light delivery


a Local somata Laser
Virally encoded
opsin
A C A C A C
B B B

Single viral injection into B Opsin expression throughout B Illumination of B cell bodies
projecting to A and C
b Recombinase- or Virally encoded Laser
promoter-dependent recombinase- or promoter-
dependent opsin
A C A C A C
B B B

Opsin expression only in neurons Illumination of B cell bodies and


Single viral injection into modulation of recombinase-
expressing recombinase or
mixed population of neurons in B or promoter-expressing cells
with active promoter in B
c Projection
Laser
Virally encoded
opsin
A C A C A C
B B B

Single viral injection into B Opsin expression throughout B Illumination of B axons in A but not C.
Corresponding cell bodies in B may
be activated.
d Projection
termination Virally encoded Virally encoded Laser Laser
lectinrecombinase recombinase-
fusion dependent opsin or
A C A C A C
B B B

Double viral injection into B and A. Opsin expression in B neurons Illumination of B cell bodies
Recombinase expressed in A moves that project to A (also achievable or B axons in A without
transcellularly to cells in B. with axon-transducing viruses) direct modulation of C

e Combinatorial Laser 1
local somata Mixture of virally
encoded opsins Laser 2
A C A C A C
B B B

Mixed viral injection with Opsin expression in B cell bodies Precisely temporally separable illumination
spectrally separated opsins into according to viral promoters or of mixed neuronal populations in B with
mixed population of neurons in B recombinase-dependent expression two colours of light
f Combinatorial
Laser 1
projection Independent
virally encoded
opsins Laser 2
A C A C A C
B B B

Viral injection of spectrally Opsin expression in A and C cell Illumination of mixed neuronal
separated opsins into A and C bodies according to viral promoters projections in B activate independent
or recombinase activity axons from A and C

256 | APRIL 2012 | VOLUME 13 www.nature.com/reviews/neuro


Nature Reviews | Neuroscience
2012 Macmillan Publishers Limited. All rights reserved
REVIEWS

Figure 2 | Targeting strategies with optogenetic tools invivo. a | Neuronal cell consideration, it is important to understand where
bodies can be directly stimulated by injecting a viral vector into the target region and long-term fear memories are stored and how they can
implanting a local light-delivery device in the same region. b | Specific expression of the be most effectively disrupted or reconfigured.
transgene in defined cell populations can be achieved by including cell-type-specific It has long been suggested that the hippocampus is
promoters within the viral vector or by injecting a recombinase-dependent virus into an important for the encoding of contextual fear 8891 but
animal that is engineered to express a recombinase (such as Cre) in particular cell types.
that long after consolidation (that is, in the remote
c | The optogenetic tool can be targeted to axonal projections by injecting the virus at
the location of neuronal cell bodies and delivering light to the target region harbouring phase many weeks, months or years later) the memory
opsin-expressing processes. d | In projection termination labelling, cells are targeted by is no longer stored in the hippocampus and is instead
virtue of their synaptic connectivity to the target region, probably excluding axons that maintained in a distributed neocortical network such
are simply passing through the area. In the example shown, transcellular labelling is as in the anterior cingulate cortex 9296. However, in a
achieved using a recombinase-dependent system. The synaptic target site is injected recent study in which eNpHR3.0 was expressed in glu-
with a virus expressing Cre that is fused to a transneuronal tracer (such as a lectin), and tamatergic pyramidal neurons in the CA1 region of the
the cell body region is injected with a Cre-dependent virus. This results in cells that hippocampus, it was shown that the hippocampus is
project to the Cre-injected area becoming light sensitive. Similar effects can be obtained indeed important for the expression of even remote fear
using retrograde viruses (those that transduce the axon terminal), such as rabies or memories. Neocortical networks are also important in
herpes simplex viruses (HSVs), although these approaches do not enable control over the
this remote (long-term) phase and can be recruited to
postsynaptic cell type. e,f | Combinatorial manipulations at either neuronal somata (e) or
projections (f) can be achieved with two different optogenetic tools that have participate more heavily when the hippocampus is dys-
well-separated activation spectra (responding to different wavelengths of light) and by functional97. In other words, even for remote fear mem-
using a light-delivery tool to merge multiple wavelengths of light. Figure is modified, with ories, the memory trace is stored in several locations
permission, from REF.6 (2011) Elsevier. that may be redundant, that may work especially hard
to compensate for each other when needed and that
therefore could require a number of simultaneous dis-
studies have implicated the amygdala in the acquisi- tinct methods and approaches to resolve pathological
tion and expression of conditioned fear 71,7381, the forms ofrecall.
intricate microcircuits in the amygdala have been In summary, applying optogenetic tools to the study
difficult to causally dissect with traditional manipula- of fear and anxiety behaviours in rodents has yielded
tions. Although optogenetic animal studies do not yet valuable and in many cases surprising results that
directly address panic disorder or phobias per se, by would have been difficult or impossible to obtain with
using optogenetic techniques to target protein kinase other methods. Fear conditioning is an excellent target
C (PKC)-expressing neurons in a subnucleus of the for the early application of optogenetics to brain disease
lateral division of the CeA, researchers have recently as it involves a simple, robust behavioural paradigm for
found an inhibitory microcircuit in the CeA that gates which the underlying neural circuitry has been exten-
the expression of conditioned fear82. Another study sively characterized and is therefore ideal for validating
defined subpopulations of neurons in the CeA that are novel techniques as well as advancing and refining our
involved in the expression and generalization of con- understanding of an emotional state that is perturbed
ditioned fear 83 (FIG.3). These studies have highlighted in many diseases.
the synergistic value of genetic and spatial targeting of
optogenetic control by combining the focal injection Circuitry of addiction
of opsin-bearing viruses into the amygdala for spatial Drug addiction is a chronic, relapsing condition charact
resolution with genetic targeting strategies (for exam- erized by compulsive drug seeking and substance use
ple, targeting PKC cells within the amygdala) for cell despite harmful consequences98100. Addiction has been
type resolution. This approach has enabled researchers proposed to hijack the brains natural reward sys-
to determine the role of a particular cell type in the tem99102; therefore, understanding the neural circuitry
control of a symptom related to psychiatric disease in mediating reward processing may be crucial for under-
animalmodels. standing the pathophysiology of addiction. Although
PTSD is a class of pervasive anxiety disorder that it has long been known that the mesolimbic dopamine
can occur following a traumatic experience under system is involved in reward processing 103107 and that
specific conditions (for example, involving subjective the NAc is critically involved in both reward processing
helplessness) and is remarkably debilitating owing to and other addiction-related behaviours100,101,105,108110,
its chronic influence on many aspects of social and the mechanistic processes and neural codes mediating
occupational functioning and its resistance to treat- these behaviours have been incompletely understood.
ment or extinction8486. Indeed, an animal model of the The motivation to understand these mechanistic
processes that may be dysfunctional in PTSD is fear processes spurred the development and application of
extinction, in which fear responses are gradually elimi- new optogenetic targeting strategies. Promoters that
nated once the aversive stressor is no longer presented could drive the expression of opsin genes specifically
with the previously associated contextual stimulus86,87. in dopamine neurons are too large to be packaged
An intriguing idea is that the fear associations in PTSD together with the microbial opsins into conventional
are pervasive and intractable to treatment because viral vectors while maintaining functionality. However,
they are stored in several locations and circuits as mentioned above, highly selective Cre-dependent
throughout the brain, with many independent viral vectors have been developed29 that enable opsins
and potent memory traces 84,86,87. Taking this into to be expressed exclusively in Cre+ cells. Notably, this

NATURE REVIEWS | NEUROSCIENCE VOLUME 13 | APRIL 2012 | 257

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

a b e

CeL
On
LA
O

CeM

Freezing

c d

CeL
PKC
On

PKC+ O ?
CeL CeL
PKC On
BLA
O
PKC+ CeM
BLA
CeM CeM
PAG

GABA-releasing terminals Glutamate-releasing terminals 500 m

Figure 3 | Functional dissection of amygdala microcircuitry using an integrated approach involving optogenetic
tools. ad | Four recent papers69,56,82,83 have used optogenetic tools to dissect subpopulations of amygdala
Nature Reviewsneurons or
| Neuroscience
projections in fear and anxiety studies. The diagrams show connectivity but are not intended to depict ultrastructural
anatomy. a | In one study69, adeno-associated virus (AAV) 2/1 serotype was used to express channelrhodopsin 2 (ChR2) in
glutamate neurons in the lateral amygdala (LA), and LA somata illumination (in lieu of shock) was paired with a conditioned
stimulus to produce fear responses. This showed that activation of LA neurons produced unconditioned fear responses
(freezing), which when paired with a stimulus was able to support stimulus-evoked freezing. b,c | Recent studies82,83
identified two interesting subpopulations of neurons in the centrolateral nucleus of the amygdala (CeL). In the experiment
shown in b, a recombinant AAV (rAAV) was used to express ChR2 in the CeL and illumination of the centromedial nucleus of
the amygdala (CeM) was used to produce fear responses83 (b). When the conditioned stimulus was presented, two
populations of CeL neurons were identified: CeL On cells showed excitation, whereas CeL Off cells were inhibited. In the
experiment shown in c, AAV2/5 expressing ChR2 was injected into the CeL of protein kinase C (PKC)::GluCl-iresCre
mice82 to show that PKC-expressing neurons inhibited periaqueductal grey (PAG)-projecting CeM neurons (c). PKC+ cells
corresponded to CeL Off cells. d | Another study56 examined amygdala function in the context of unconditioned anxiety
rather than conditioned fear. In this study, basolateral amygdala (BLA) neurons were transduced with an AAV expressing
ChR2, and it was shown that activating BLACeL projections reduced anxiety-related behaviours, whereas activating the
BLA cell bodies without specificity for projection target increased anxiety-related behaviours. This study also demonstrated
the opposite effects of BLACeL projections using optogenetic inhibition. However, this study did not determine whether
BLA neurons provided monosynaptic excitatory input to a particular subpopulation of CeL neurons. e | Information from
these four studies, synthesized to display certain current optogenetically obtained knowledge about amygdala
microcircuitry that is causally involved in behaviour. Although many questions remain, these studies show the advantages of
integrating optogenetic tools with molecular, electrophysiological, pharmacological and imaging techniques in the context
of behaviour relevant to psychiatric disease.

strategy was used to target ChR2 to dopamine neu- a paradigm that has been used to assay drug reward-
rons in the ventral tegmental area (VTA) to show that related behaviours111,112. The use of optogenetics with
phasic, but not tonic, stimulation of VTA dopamine CPP has enabled researchers to identify the functional
neurons at frequencies that reliably produced NAc contributions of distinct neural substrates in the NAc
dopamine transients was sufficient to support CPP31, in reward-related behaviours. Distinct populations of

258 | APRIL 2012 | VOLUME 13 www.nature.com/reviews/neuro

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

place preference50, and acute optogenetic activation of


defined G protein-coupled receptor pathways in the
NAc supported CPP4 (FIG.4).
To better understand the circuits underlying reward-
3 seeking behaviour, a series of studies have examined
VTA
whether optogenetic activation of specific brain areas
NAc and neural pathways can serve as rewards to be sought
in their own right. These studies draw on a long his-
2 BLA
tory of experiments that showed that animals would
perform an operant response (usually a lever press) to
deliver small amounts of electrical current to specific
brain regions114, a behaviour that is termed intracranial
self-stimulation (ICSS). Although such experiments
served as a powerful demonstration that reward could
be triggered entirely within the CNS, the heterogene-
ous neural populations activated by this technique
precluded definitive knowledge about which cell types
and neural projections could sustain ICSS behaviour.
MSN
D2R Two recent studies used optogenetics to establish causal
1 roles for specific cell types in ICSS. Selectively activating
ChAT excitatory projections from the BLA to the NAc
4 (defined by a projection-specific targeting strategy)
produced robust self-stimulation, whereas stimulation
of the same cell bodies did not 61. Moreover, this self-
stimulation of BLANAc projections was dependent
on D1 dopamine receptor (D1R) signalling 61 (FIG.4).
Optogenetic self-stimulation of dopaminergic neu-
MSN rons in the VTA has also been demonstrated37,52 (FIG.4).
D1R
2 Together, these findings provide a greater understand-
ing of the precise activity patterns in defined cells or
projections that underlie processes that are thought to
become pathological in states of substance dependence
orabuse.
We anticipate that optogenetic tools will be lever-
aged to test ideas that are based on indirect evidence.
For example, GABAergic neurons in the VTA are
Figure 4 | Optogenetic dissection of limbic circuits in the context of
reward-seeking behaviour. A schematic diagram (top panel),
emerging as a new target for addiction research because
Natureshowing
Reviewskey neural
| Neuroscience
projections involved in reward, and an expanded view of intra-accumbens microcircuitry opioids act to reduce GABAergic suppression of VTA
(bottom panel; numbers indicate independent findings). The diagram shows connectivity dopamine neurons115. With the availability of trans-
but is not intended to depict ultrastructural anatomy. Cholinergic interneurons genic mouse lines selectively expressing Cre, direct
expressing choline acetyltransferase (ChAT) in the nucleus accumbens (NAc) modulate manipulation of VTA GABA neurons can be tested
the activity of medium spiny neurons (MSNs) and modulate the ability of the animal to in assays of hedonic and reward-seeking behaviours.
develop cocaine-conditioned place preference (1)50. Mice will readily work (nose poke) to Relative to fear-conditioning, reward-related behav-
receive illumination of channelrhodopsin 2 (ChR2)-expressing basolateral amygdala iours may be more sensitive to motivational state.
(BLA) axon terminals in the NAc (2). This self-stimulation behaviour is D1 dopamine However, previous invivo electrophysiological record-
receptor (D1R)-dependent, and does not occur when ChR2expressing prefrontal cortex
ings performed throughout the corticolimbic system
(PFC) axon terminals in the NAc are illuminated61. It has been shown that activation of
dopaminergic neurons in the ventral tegmental area (VTA) can support operant
during the acquisition116119, maintenance106,108,110,120122,
responding in both rats and mice52, and in rats, illumination of tyrosine hydroxylase and extinction 123,124 and reinstatement 123,125 of reward-
ChR2expressing axon terminals in the NAc also supports operant responding (3)37. The related behaviours have identified a rich landscape of
activation of D1R- or D2Rexpressing NAc neurons shows differential effects on cocaine- neural correlates, and have set the stage for optogenetic
conditioned place preference (4)113. interrogation in the pursuit of causality.
The strong invivo electrophysiological tradition
in this field is also supported by studies that used
neurons in the NAc that express different dopamine intracranial pharmacological manipulations to iden-
receptor subtypes differentially modulate cocaine place tify circuits relevant to addiction and reward-seeking
preference; activating D1expressing neurons enhances behaviours. Thus, looking towards the further integra-
cocaine CPP, whereas activating D2expressing neu- tion of optogenetics with high-speed readouts for neu-
rons suppresses cocaine CPP113. In related work that ral activity and neurochemical signals126,127, the field of
also combined optogenetics with the use of CPP, reward-related behaviour represents a fertile proving
cholinergic neurons in the NAc were shown to be cru- ground for technological synergy set against a back-
cially involved in modulating cocaine reward-related drop of sophisticated behavioural theory.

NATURE REVIEWS | NEUROSCIENCE VOLUME 13 | APRIL 2012 | 259

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

Circuitry of depression An intrinsic obstacle in studying the neural circuits


Although up to 13% of the population will experience underlying depression is that the constellation of symp-
clinically relevant major depression at some point dur- toms that define clinical depression is not as readily
ing life64,128, the pathophysiological underpinnings of modelled in animals as other diseases. For example,
depression and other mood disorders are poorly under- how can motivation be assessed independently from
stood. Currently available antidepressant medications locomotion, and how can despair and helplessness be
are often ineffective and even when effective must gen- reliably assayed in animals? In contrast to addiction
erally be taken for 46weeks to show improvements research in which the disease is defined by the behav-
in patients 129132. Brain stimulation treatments that iour, in depression the disease state is mainly defined by
are milder and more targeted than electroconvulsive the subjective experience of the patient. This is a major
therapy are also being actively explored; for example, challenge with which depression researchers continue
high-frequency electrical stimulation of the region near to wrestle to advance our understanding of this severely
the subgenual cingulate gyrus may improve depres- debilitating and common psychiatric disease. It may be
sive symptoms in treatment-resistant patients133,134. effective to focus on the symptoms that have measur-
Although this breakthrough finding sparked hope of able correlates in animals (such as assays for changes in
an improved understanding of the neural substrates motivated behaviours in the absence of gross locomo-
of depression, the mechanisms of this phenomenon tor effects, and hedonic behaviour changes). Despite
are still poorly understood; for example, it is not the practical challenges in using optogenetic techniques
clear whether high-frequency electrical stimulation to dissect the neural underpinnings of depression and
exerts antidepressant effects by directly influencing other mood disorders, the immense potential benefit
axons passing through this area or local cell bodies, or to the broader community of this reverse translational
whether the net effect that is causal in symptom remedi- approach warrants a proliferation of work in thisfield.
ation is increasing, decreasing or otherwise influencing
local cortical neural electrical activity. Circuitry of autism and schizophrenia
In an attempt to better understand the underlying Autism and schizophrenia also present major experi-
mechanisms of deep brain stimulation (DBS) treatments mental challenges for identifying neural circuits
for depression better, optogenetic techniques have altered in states of disease because of the heterogeneity
been used to target cell bodies in the prefrontal cor- of symptoms that each nominally unitary condition
tex of mice. Mice with a depression-related phenotype encapsulates. Patients with autism and schizophrenia
showed an antidepressant-like response to illumination display diverse and variable arrays of symptoms that
of medial prefrontal cortical neurons expressing ChR2 make the search for a single cause a daunting and pos-
(REF.135). Although this study used a more targeted sibly unrealistic goal158161. At the surface level, these
method than electrical stimulation, limiting activation diseases share certain signs and symptoms, such as
to local cell bodies without activating fibres of passage, social dysfunction, although the experienced physician
new optogenetic tools allow even greater specificity. will notice that the social behaviours appear to be quite
For example, because the prefrontal cortex projects different, even in dysfunction, in these two diseases.
to many regions, including the dorsal raphe, the NAc, Schizophrenia symptoms include disorganized
amygdala and other structures proposed to be involved speech and behaviour, as well as hallucinations, delu-
in mood regulation, it will be particularly useful to sions and negative symptoms such as impaired social
examine these targets in a pathway-specific manner function162,163. This debilitating disease has been cor-
using projection targeting approaches. In addition, the related with perturbations in the balance of excitation
use of cell-type-specific promoters or Cre-dependent and inhibition and with altered oscillations of cortical
targeting strategies would allow increased cellular, as neural networks164, both of which may in theory lead
well as regional, specificity. to a breakdown in the transmission or processing of
Depression has been linked to many neural path- neural information. Although animal models of schizo-
ways, including corticolimbic134, dorsal raphe136138, hip- phrenia are still being optimized, one of the few reliable
pocampal139142, amygdalar 143146,6568, striatal147149 and micro-anatomical abnormalities in psychiatric disease
mesolimbic dopamine130,150152 circuits. Furthermore, is the reduced number and functionality of neocortical
depression has been linked to distinct neuromodula- parvalbumin neurons165,166, and therefore optogenetic
tory systems and receptors, including serotonin153,154, approaches to the circuitry in question have included
noradrenaline155 and dopamine130,150152,155,156. Most cur- targeting these neurons in PV::Cre mouse lines 53,54.
rently available antidepressants modulate monoamines, These studies have revealed the roles of parvalbumin
globally modulating synaptic neurotransmission using neurons in modulating gamma rhythms and informa-
serotonin, dopamine and/or noradrenaline. Given the tion processing in neocortex that may help to lay the
diverse distribution and functionality of these recep- groundwork for a deeper understanding of information
Deep brain stimulation tors, increasing the specificity of drug targets could processing deficits in schizophrenia53,54.
(DBS). Continuous therapeutic dramatically improve drug efficacy or reduce side Autism spectrum disease is characterized by impair-
electric stimulation of effects157. Future development and implementation of ment in social behaviour and communication as well
subcortical areas at high
frequencies (~130Hz) using
optogenetic tools may inspire drug development with as stereotyped, repetitive movements159,160,167. Autism
chronically implanted enhanced precision to provide more efficacious anti is co-morbid with neurological disorders such as
electrodes. depressants with fewer side effects. epilepsy 168,169, and is also linked to anxiety and mood

260 | APRIL 2012 | VOLUME 13 www.nature.com/reviews/neuro

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

disorders158,170. The complexity and variability of symp- therapy have been unclear and highly controversial.
toms in autism have made it particularly difficult to Selective optogenetic control of afferent fibres (but not
study, but one theory has been that imbalance in cel- selective control of local somata) in the STN was shown
lular excitation and inhibition (excitationinhibition to have profound therapeutic effects on motor behav-
imbalance) may be causative171173. Optogenetic tools iours in a hemi-parkinsonian rat model103 (FIG.5). These
offered the ability to empirically test this idea by ena- data suggested a model for DBS treatment in which
bling the induction of excitationinhibition imbalance. white matter tracts or axonal pathways are the most
The expression of a SSFO, which produces subthreshold effective direct target of control. The reasons for the
membrane potential changes that last many minutes, in efficacy of control by this means could include the fact
glutamatergic prefrontal cortical neurons allowed the that recruitment of large or asymmetric neural struc-
level of cortical excitation to be elevated48. This resulted tures with a point source of energy such as an electrode
in an increase in rhythmic activity in the gamma band or an optical fibre is most efficient if spatially local-
(3080Hz) a trait linked to autism174 and also vir- ized incoming axon tracts are the direct initial target.
tually abolished unconditioned social behaviour 48. To This concept carries direct implications for electrode
determine whether this effect was related to an imbal- contact positioning in DBS for depression as well as
ance in the activity of excitatory and inhibitory cells, a Parkinsons disease. Alternatively (or additionally), it
novel red-shifted opsin developed for the purpose of may be that the flow of activity between brain regions
combinatorial excitation was used to partially rescue that can be targeted at the level of axon tracts (rather
the phenotype by simultaneously increasing the drive than neural coding by somata perse within a region)
of both excitatory and inhibitorycells48. is the most functionally relevant endophenotype for
An imbalance in excitation and inhibition may also neuropsychiatric disease symptomatology.
contribute to other psychiatric disorders, such as anxi- Pathway-specific optogenetic approaches in com-
ety and depression56, and may give rise to interesting bination with whole-cell recordings in a corticotha-
candidate endophenotypes in other circuits, such as lamic circuit linked to epilepsy revealed a pathological
rhythmic oscillations (for example, gamma oscilla- generation of aberrant oscillations relevant to seizure
tions) and altered activity percolation through the activity 169. Optogenetic activation of neurons in the
circuit. Indeed, given the highly overlapping circuits retrotrapezoid nucleusparafacial respiratory group
that are involved in psychiatric disorders such as anxi- induced active expiration and regulated the rhythm of
ety, depression, addiction, schizophrenia and autism, respiration181. It may be worth noting that application
it is possible that excitation and inhibition imbalances of optogenetic approaches can also further the study of
throughout the brain could contribute to the high rate stem cell-based interventions for Parkinsons disease191;
of co-morbidity among these disease states156,158,162,175178, optogenetic targeting of grafted stem-cell-derived
and optogenetic tools now offer the capability to test dopamine neurons has shown that opsin-expressing
these fascinating and potentially unifying theories. grafted cells can be functionally integrated into net-
works of wild-type mouse striatum 168. The uncondi-
Circuitry of neurological disorders tioned modulation of feeding behaviour was shown
Optogenetic approaches have been applied to funda- to be under the control of two functionally opposing
mental research questions in a variety of neurological subpopulations of neurons in the hypothalamus57. An
disorders such as Parkinsons disease51,55, epilepsy 168,169, elegant examination of aggressive behaviour demon-
blindness due to neuronal loss179,180, failure of respira- strated that attacks could be evoked with optogenetic
tion181,182 and neuropsychiatric sleep disorders25. Our activation of a specific subset of ventromedial hypo-
basic understanding of motor circuitry has also been thalamic neurons192. Lastly, restoration of vision by
enhanced by optogenetic investigations; for example, optogenetic activation of cones was shown to rescue
it had long been suggested that the basal ganglia regu- the blindness associated with retinitis pigmentosa179,180.
late movement through the balance of two pathways,
the direct pathway, which promotes locomotion, Conclusion
and the indirect pathway, which inhibits locomo- We have provided here a brief summary of some
tion183185. Selective targeting of each of these pathways recent investigations relevant to the study of neu-
was made possible by Cre-dependent optogenetic ropsychiatric disease that used optogenetic tools. In
targeting of D1R- or D2R-expressing cells51, thus pro- these cases, the precision of optogenetics has pro-
viding the first direct empirical confirmation of this vided major experimental leverage193 and has led to
long-standingtheory. insights into neural circuit function and dysfunction.
Parkinsons disease, a neurodegenerative disease, is Although the impact of these recent investigations has
characterized by rigidity, tremor, postural instability already been substantial, there remains much work
and slow movement 186,187. Although DBS in the subgen- to do; for example, the application of optogenetics to
ual cingulate cortex has been used in humans to treat non-human primates is still in its infancy 24,27,194,195,
depression133, DBS in the subthalamic nucleus (STN), and many disease states and symptom clusters remain
a part of the basal ganglia circuit, and in other targets unexplored. We anticipate several major areas of
has shown remarkable therapeutic effects for treatment optogenetic tool advancement in the future, from tool
of the motor impairments associated with Parkinsons development to scientific application, mainly driven
disease188190. However, the cellular mechanisms of this by demand in thefield.

NATURE REVIEWS | NEUROSCIENCE VOLUME 13 | APRIL 2012 | 261

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

M1

Striatum
D2 D1
GP
SNr
STN

GABA-releasing terminals
STN cells upon aerent activation Glutamate-releasing terminals

HFS
Striatal illumination in D1R::Cre or D2R::Cre mice
50 V
4s
LFS

50 V
4s 12 D1 20 D2

Hz

Hz
50 V 0 0
50 V 1 0 1 2
0.5 s 5 ms 1 0 1 2
Time (s) Time (s)

Figure 5 | Functional mapping of basal ganglia circuitry using optogenetics in the context of Parkinsons disease.
A schematic diagram (top panel) shows key neural projections that are involved in parkinsonianNature Reviews
behaviour | Neuroscience
and treatment.
Data in the bottom left panel are from a study that used a constitutively expressing channelrhodopsin 2 (ChR2) mouse line
(Thy1::ChR2) to identify a mechanistic explanation for the therapeutic effects of deep brain stimulation (DBS) 55. By illuminating
and recording in the subthalamic nucleus (STN), this paper showed that afferent fibres entering the STN, rather than local cell
bodies themselves, are likely to be the direct target of DBS in the correction of parkinsonian motor activity. High-frequency
stimulation (HFS) of the afferent fibres into STN potently silenced the structure as shown and reversibly abolished the
parkinsonian symptoms. By contrast, low-frequency stimulation (LFS) of the afferents simply added spikes on top of
endogenous spikes and worsened parkinsonian symptoms. Data in the bottom right panel are from a study that used a
Cre-dependent adeno-associated virus (AAV) to selectively express ChR2 in either D1 dopamine receptor (D1R)::Cre or
D2R::Cre mice to examine the differential contributions of the direct and indirect pathways with respect to motor output.
Activation of D1Rexpressing neurons silenced local basal ganglia activity and increased ambulation, whereas activation of
D2Rexpressing neurons increased this activity and enhanced immobile or bradykinetic (slow) behaviour51. Black bars indicate
the duration of illumination. The bottom left panel is reproduced, with permission, from REF. 55 (2009) American Association
for the Advancement of Science. The bottom right panel is reproduced, with permission, from REF. 51 (2010) Macmillan
Publishers Ltd. All rights reserved. GP, globus pallidus; M1, primary motor cortex; SNr, substantia nigra pars reticulata.

With respect to developments in opsin engineer- variants. Improved or expanded recombinase strategies
ing, despite the rapidly proliferating library of opsins, are well within reach, and would facilitate circuit-level
there remain areas that require increased attention. For investigations. We have steadily improved the expres-
example18, we anticipate the engineering of blue-shifted sion of opsins at the membrane, but further exploration
hyperpolarizing opsins with narrower activation wave- in this area may produce targeting strategies that allow
length spectra to allow for enhanced combinatorial neu- selective opsin expression in subcellular compartments
ronal inhibition experiments21. Another much needed such as dendrites, soma or axon terminals, thereby
development is a hyperpolarizing SFO to allow sustained increasing the level of precision196. Currently, we are
inhibition of neurons without requiring constant illumi- able to selectively express opsins in cells with a given
nation, which can cause heating and may be impractical genetic phenotype or marker, but methods to selec-
for chronic inhibition experiments. In addition to light- tively exclude opsin expression in cells with a given
sensitive pumps and channels, we anticipate the contin- genetic identity will also beuseful.
ued expansion of the OptoXR family 4, as light-sensitive In addition, methods of light delivery will require
domains are being added to an increasing number of continual improvement. The optical fibre provides
receptor and even intracellular signalling proteins so that a small zone of illumination and, rather than opsin
optogenetics will expand to the study of cell signalling in expression, the illumination pattern may often be limit-
addition to the study of neural activity. ing. Many experiments may call for broader volumes of
The development of improved targeting strate- illumination (to facilitate achieving behavioural effects
gies on both a cellular and subcellular level is just as from optogenetic manipulations in larger species such as
important as the development of new opsins and opsin monkeys). We also anticipate continued improvement

262 | APRIL 2012 | VOLUME 13 www.nature.com/reviews/neuro

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

in the speed and precision of single-cell activation anxiety, depression, addiction, schizophrenia and
through refinement of the ability to visualize and selec- autism. Variation in precise symptomatology from dis-
tively illuminate individual cells to allow the playback ease to disease could be more closely linked to variation
of specific neuronal population codes. The creation of a in the role of the affected circuit than to fundamentally
method to simultaneously perform optogenetic activa- distinct principles of the pathologically altered neural
tion and achieve calcium imaging at a single-cell or even activity propagation. In particular, given the high rate
dendritic-spine resolution in a freely moving mammal of co-morbidity among the various mental illnesses
would also be an enormous advance for thefield. and the shared symptomatology between individual
It is intriguing to speculate that a very specific (and diseases, identification of such simplifying themes
now testable) neural circuit dysfunction (such as an and unifying theories by optogenetic or other means is
imbalance in excitation and inhibition) could be caus- one of the most pressing needs and exciting avenues of
ally involved in multiple psychiatric diseases including research into neurological and psychiatricdisease.

1. Deisseroth, K. Optogenetics. Nature Methods 8, 19. Chow, B.Y. etal. High-performance genetically 36. Wang, J. etal. Integrated device for combined optical
2629 (2011). targetable optical neural silencing by light-driven neuromodulation and electrical recording for chronic
2. Deisseroth, K. etal. Next-generation optical proton pumps. Nature 463, 98102 (2010). invivo applications. J.Neural Eng. 9, 016001 (2012).
technologies for illuminating genetically targeted brain 20. Gunaydin, L.A. etal. Ultrafast optogenetic control. 37. Witten, I.B. etal. Recombinase-driver rat lines: tools,
circuits. J.Neurosci. 26, 1038010386 (2006). Nature Neurosci. 13, 387392 (2010). techniques, and optogenetic application to dopamine-
3. Zhang, F. etal. The microbial opsin family of 21. Mattis, J. etal. Principles for applying optogenetic mediated reinforcement. Neuron 72, 721733
optogenetic tools. Cell 147, 14461457 (2011). tools derived from direct comparative analysis of (2011).
4. Airan, R.D., Thompson, K.R., Fenno, L.E., microbial opsins. Nature Methods 9, 159172 38. Lee, J.H. etal. Global and local fMRI signals driven by
Bernstein, H. & Deisseroth, K. Temporally precise (2011). neurons defined optogenetically by type and wiring.
invivo control of intracellular signalling. Nature 458, An empirical comparison of most existing microbial Nature 465, 788792 (2010).
10251029 (2009). opsin-derived optogenetic tools under matched 39. Peterlin, Z.A., Kozloski, J., Mao, B.Q., Tsiola, A. &
This study developed the OptoXRs (photosensitive conditions, including the development of several Yuste, R. Optical probing of neuronal circuits with
G protein-coupled receptors based on vertebrate new opsin variants. calcium indicators. Proc. Natl Acad. Sci. USA 97,
opsin genes) for mammalian invivo use and 22. Iwai, Y., Honda, S., Ozeki, H., Hashimoto, M. & 36193624 (2000).
showed that light-activated intracellular Hirase, H. A simple head-mountable LED device for 40. Shoham, S. Optogenetics meets optical wavefront
signalling cascades could support conditioned chronic stimulation of optogenetic molecules in freely shaping. Nature Methods 7, 798799 (2010).
place preference. moving mice. Neurosci. Res. 70, 124127 (2011). 41. Petreanu, L., Huber, D., Sobczyk, A. & Svoboda, K.
5. Boyden, E.S., Zhang, F., Bamberg, E., Nagel, G. & 23. Schneider, M.B., Gradinaru, V., Zhang, F. & Channelrhodopsin-2assisted circuit mapping of long-
Deisseroth, K. Millisecond-timescale, genetically Deisseroth, K. Controlling neuronal activity. Am. range callosal projections. Nature Neurosci. 10,
targeted optical control of neural activity. Nature J.Psychiatry 165, 562 (2008). 663668 (2007).
Neurosci. 8, 12631268 (2005). 24. Diester, I. etal. An optogenetic toolbox designed for 42. Petreanu, L., Mao, T., Sternson, S.M. & Svoboda, K.
The initial demonstration of single-component primates. Nature Neurosci. 14, 387397 (2011). The subcellular organization of neocortical excitatory
optogenetics using a microbial opsin gene (in this 25. Adamantidis, A.R., Zhang, F., Aravanis, A.M., connections. Nature 457, 11421145 (2009).
case, a channelrhodopsin). Deisseroth, K. & de Lecea, L. Neural substrates of 43. Adesnik, H. & Scanziani, M. Lateral competition for
6. Yizhar, O., Fenno, L.E., Davidson, T.J., Mogri, M. & awakening probed with optogenetic control of cortical space by layer-specific horizontal circuits.
Deisseroth, K. Optogenetics in neural systems. Neuron hypocretin neurons. Nature 450, 420424 (2007). Nature 464, 11551160 (2010).
71, 934 (2011). The first application of a microbial opsin to show a 44. Han, X. & Boyden, E.S. Multiple-color optical
A comprehensive practical overview describing the behavioural change in a mammal (gain of function, activation, silencing, and desynchronization of neural
implementation of optogenetic tools in neural using a channelrhodopsin). activity, with single-spike temporal resolution. PLoS
systems. 26. Grossman, N. etal. Multi-site optical excitation using ONE 2, e299 (2007).
7. Fenno, L., Yizhar, O. & Deisseroth, K. The development ChR2 and micro-LED array. J.Neural Eng. 7, 016004 45. Zhao, S. etal. Improved expression of halorhodopsin
and application of optogenetics. Annu. Rev. Neurosci. (2010). for light-induced silencing of neuronal activity. Brain
34, 389412 (2010). 27. Bernstein, J.G. etal. Prosthetic systems for Cell Biol. 36, 141154 (2008).
8. Carter, M.E. & de Lecea, L. Optogenetic investigation therapeutic optical activation and silencing of 46. Gradinaru, V., Thompson, K.R. & Deisseroth, K.
of neural circuits invivo. Trends Mol. Med. 17, genetically-targeted neurons. Proc. Soc. Photo Opt. eNpHR: a Natronomonas halorhodopsin enhanced for
197206 (2011). Instrum. Eng. 6854, 68540H (2008). optogenetic applications. Brain Cell Biol. 36,
9. Aravanis, A.M. etal. An optical neural interface: 28. Sparta, D.R. etal. Construction of implantable optical 129139 (2008).
invivo control of rodent motor cortex with integrated fibers for long-term optogenetic manipulation of 47. Lin, J.Y., Lin, M.Z., Steinbach, P. & Tsien, R.Y.
fiberoptic and optogenetic technology. J.Neural Eng. neural circuits. Nature Protoc. 7, 6851268523 Characterization of engineered channelrhodopsin
4, S143S156 (2007). (2011). variants with improved properties and kinetics.
10. Kravitz, A.V. & Kreitzer, A.C. Optogenetic 29. Atasoy, D., Aponte, Y., Su, H.H. & Sternson, S.M. Biophys. J. 96, 18031814 (2009).
manipulation of neural circuitry invivo. Curr. Opin. A FLEX switch targets Channelrhodopsin2 to multiple 48. Yizhar, O. etal. Neocortical excitation/inhibition
Neurobiol. 21, 433439 (2011). cell types for imaging and long-range circuit mapping. balance in information processing and social
11. Lima, S.Q. & Miesenbck, G. Remote control of J.Neurosci. 28, 70257030 (2008). dysfunction. Nature 477, 171178 (2011).
behavior through genetically targeted 30. Kuhlman, S.J. & Huang, Z.J. High-resolution labeling The introduction of two new opsin variants, C1V1
photostimulation of neurons. Cell 121, 141152 and functional manipulation of specific neuron types in and SSFO, for combinatorial optogenetic excitation
(2005). mouse brain by Cre-activated viral gene expression. invivo. The study showed the importance of altered
12. Nagel, G. etal. Channelrhodopsin1: a light-gated PLoS ONE 3, e2005 (2008). excitation/inhibition ratios to social behaviour and
proton channel in green algae. Science 296, 31. Tsai, H.C. etal. Phasic firing in dopaminergic neurons cognition.
23952398 (2002). is sufficient for behavioral conditioning. Science 324, 49. Berglund, K., Kuner, T., Feng, G. & Augustine, G.J.
13. Nagel, G. etal. Channelrhodopsin2, a directly light- 10801084 (2009). Imaging synaptic inhibition with the genetically
gated cation-selective membrane channel. Proc. Natl The initial behavioural application of the encoded chloride indicator clomeleon. Cold Spring
Acad. Sci. USA 100, 13940 13945 (2003). double-floxed inverted open reading frame tool Harb. Protoc. 2011, 14921497 (2011).
14. Zhang, F. etal. Red-shifted optogenetic excitation: a enabling selective opsin expression in neurons 50. Witten, I.B. etal. Cholinergic interneurons control
tool for fast neural control derived from Volvox carteri. positive for Cre recombinase. This is used in the local circuit activity and cocaine conditioning. Science
Nature Neurosci. 11, 631633 (2008). TH::Cre mouse to demonstrate the hedonic 330, 16771681 (2010).
15. Berndt, A. etal. High-efficiency channelrhodopsins for properties of phasic dopamine neuron firing. The first behavioural loss-of-function in mammals
fast neuronal stimulation at low light levels. Proc. Natl 32. Arenkiel, B.R. etal. In vivo light-induced activation of achieved using optogenetics. This study involved
Acad. Sci. USA 108, 75957600 (2011). neural circuitry in transgenic mice expressing the introduction of an inhibitory opsin into
16. Gradinaru, V. etal. Molecular and cellular approaches Channelrhodopsin2. Neuron 54, 205218 (2007). cholinergic interneurons using the double-floxed
for diversifying and extending optogenetics. Cell 141, 33. Zhao, S. etal. Cell type-specific channelrhodopsin2 inverted open reading frame tool in a choline
154165 (2010). transgenic mice for optogenetic dissection of neural acetyltransferase (ChAT)::Cre mouse to
17. Berndt, A., Yizhar, O., Gunaydin, L.A., Hegemann, P. circuitry function. Nature Methods 8, 745752 demonstrate the role of cholinergic neurons in
& Deisseroth, K. Bi-stable neural state switches. (2011). mediating cocaine-conditioned place preference.
Nature Neurosci. 12, 229234 (2009). 34. Taniguchi, H. etal. A resource of cre driver lines for 51. Kravitz, A.V. etal. Regulation of parkinsonian motor
18. Zhang, F. etal. Multimodal fast optical interrogation of genetic targeting of GABAergic neurons in cerebral behaviours by optogenetic control of basal ganglia
neural circuitry. Nature 446, 633639 (2007). cortex. Neuron 71, 9951013 (2011). circuitry. Nature 466, 622626 (2010).
The first behavioural lossoffunction using 35. Anikeeva, P. etal. Optetrode: a multichannel readout This study used optogenetics to provide empirical
optogenetics; achieved here with the for optogenetic control in freely moving mice. Nature evidence of the long-proposed effects of the direct
inhibitory optogenetic tool NpHR. Neurosci. 15, 163170 (2012). and indirect pathways in the striatum on movement.

NATURE REVIEWS | NEUROSCIENCE VOLUME 13 | APRIL 2012 | 263

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

52. Adamantidis, A.R. etal. Optogenetic interrogation of 72. LeDoux, J.E. Emotion: clues from the brain. Annu. 97. Goshen, I. etal. Dynamics of retrieval strategies
dopaminergic modulation of the multiple phases of Rev. Psychology 46, 209235 (1995). for remote memories. Cell 147, 678689
reward-seeking behavior. J.Neurosci. 31, 73. Fanselow, M.S. & Gale, G.D. The amygdala, fear, and (2011).
1082910835 (2011). memory. Ann. NY Acad. Sci. 985, 125134 (2003). This study showed that remote memory recall is
53. Sohal, V.S., Zhang, F., Yizhar, O. & Deisseroth, K. 74. Maren, S. Synaptic mechanisms of associative memory dependent on hippocampal activity, using
Parvalbumin neurons and gamma rhythms enhance in the amygdala. Neuron 47, 783786 (2005). fast-acting optogenetic methods.
cortical circuit performance. Nature 459, 698702 75. Jngling, K. etal. Neuropeptide Smediated control of 98. Koob, G.F. Drugs of abuse: anatomy, pharmacology
(2009). fear expression and extinction: role of intercalated and function of reward pathways. Trends Pharmacol.
Together with reference54, this study used GABAergic neurons in the amygdala. Neuron 59, Sci. 13, 177184 (1992).
optogenetic tools to link parvalbumin cells to 298310 (2008). 99. Wise, R.A. Drug-activation of brain reward pathways.
gamma rhythms, information transmission and 76. LeDoux, J., Cicchetti, P., Xagoraris, A. & Romanski, L. Drug Alcohol Depend. 51, 1322 (1998).
processing in the neocortex. The lateral amygdaloid nucleus: sensory interface of 100. Everitt, B.J. & Robbins, T.W. Neural systems of
54. Cardin, J.A. etal. Driving fast-spiking cells induces the amygdala in fear conditioning. J.Neurosci. 10, reinforcement for drug addiction: from actions to
rhythm and controls sensory responses. Nature 459, 10621069 (1990). habits to compulsion. Nature Neurosci. 8, 1481
663667 (2009). 77. Miserendino, M.J.D., Sananes, C.B., Melia, K.R. & (2005).
55. Gradinaru, V., Mogri, M., Thompson, K.R., Davis, M. Blocking of acquisition but not expression of 101. Robinson, T.E. & Berridge, K.C. The neural basis of
Henderson, J.M. & Deisseroth, K. Optical conditioned fear-potentiated startle by NMDA drug craving: an incentive-sensitization theory of
deconstruction of parkinsonian neural circuitry. antagonists in the amygdala. Nature 345, 716718 addiction. Brain Res. Rev. 18, 247291 (1993).
Science 324, 354359 (2009). (1990). 102. Kelley, A.E. & Berridge, K.C. The neuroscience of
This study provided a mechanistic explanation for 78. Par, D., Quirk, G.J. & Ledoux, J.E. New vistas on natural rewards: relevance to addictive drugs.
how DBS alleviates parkinsonian symptoms using amygdala networks in conditioned fear. J.Neurosci. 22, 33063311 (2002).
optogenetic techniques in animal models. J.Neurophysiol. 92, 19 (2004). 103. Schultz, W., Dayan, P. & Montague, P.R. A neural
56. Tye, K.M. etal. Amygdala circuitry mediating 79. Rodrigues, S.M., Schafe, G.E. & LeDoux, J.E. Intra- substrate of prediction and reward. Science 275,
reversible and bidirectional control of anxiety. amygdala blockade of the NR2B subunit of the NMDA 15931599 (1997).
Nature 471, 358362 (2011). receptor disrupts the acquisition but not the 104. Spanagel, R. & Weiss, F. The dopamine hypothesis of
The first optogenetic projection-specific targeting expression of fear conditioning. J.Neurosci. 21, reward: past and current status. Trends Neurosci. 22,
study of behaviour, showing that acute activation of 68896896 (2001). 521527 (1999).
neuron axon terminals can produce an opposite 80. Wilensky, A.E., Schafe, G.E., Kristensen, M.P. & 105. Berridge, K.C. & Robinson, T.E. What is the role of
effect on anxiety to that produced by activating LeDoux, J.E. Rethinking the fear circuit: the central dopamine in reward: hedonic impact, reward learning,
neuron somata. It was also the first study to show nucleus of the amygdala is required for the acquisition, or incentive salience? Brain Res. Rev. 28, 309369
axon-terminal specific inhibition using optogenetics consolidation, and expression of pavlovian fear (1998).
invivo. conditioning. J.Neurosci. 26, 1238712396 (2006). 106. Ikemoto, S. & Panksepp, J. The role of nucleus
57. Aponte, Y., Atasoy, D. & Sternson, S.M. AGRP 81. Han, J.H. etal. Selective erasure of a fear memory. accumbens dopamine in motivated behavior: a
neurons are sufficient to orchestrate feeding behavior Science 323, 14921496 (2009). unifying interpretation with special reference to
rapidly and without training. Nature Neurosci. 14, 82. Haubensak, W. etal. Genetic dissection of an reward-seeking. Brain Res. Rev. 31, 641 (1999).
351355 (2011). amygdala microcircuit that gates conditioned fear. 107. Kalivas, P.W. & Stewart, J. Dopamine transmission in
58. Hgglund, M., Borgius, L., Dougherty, K. J. & Kiehn, O. Nature 468, 270276 (2010). the initiation and expression of drug-and stress-
Activation of groups of excitatory neurons in the Together with reference83, this study identified a induced sensitization of motor activity. Brain Res. Rev.
mammalian spinal cord or hindbrain evokes new population of neurons, and identified 16, 223244 (1991).
locomotion. Nature Neurosci. 13, 246252 functional specificity in PKC-expressing neurons in 108. Ambroggi, F., Ishikawa, A., Fields, H.L. & Nicola, S.M.
(2010). a multisynaptic circuit in the context of conditioned Basolateral amygdala neurons facilitate reward-
59. Choi, J., Young, J.A.T. & Callaway, E.M. Selective fear expression. seeking behavior by exciting nucleus accumbens
viral vector transduction of ErbB4 expressing cortical 83. Ciocchi, S. etal. Encoding of conditioned fear in neurons. Neuron 59, 648661 (2008).
interneurons invivo with a viral receptor-ligand central amygdala inhibitory circuits. Nature 468, 109. Cardinal, R.N., Parkinson, J.A., Hall, J. & Everitt, B.J.
bridge protein. Proc. Natl Acad. Sci. USA 107, 277282 (2010). Emotion and motivation: the role of the amygdala,
1670316708 (2010). 84. Yehuda, R. Post-traumatic stress disorder. N.Engl. ventral striatum, and prefrontal cortex. Neurosci.
60. Li, F., Ryu, B.Y., Krueger, R.L., Heldt, S.A. & J.Med. 346, 108114 (2002). Biobehav. Rev. 26, 321352 (2002).
Albritton, L.M. Targeted entry via somatostatin 85. Charney, D.S., Deutch, A.Y., Krystal, J.H., Southwick, 110. Nicola, S.M., Yun, I.A., Wakabayashi, K.T. &
receptors using a novel modified retrovirus S.M. & Davis, M. Psychobiologic mechanisms of Fields, H.L. Firing of nucleus accumbens neurons
glycoprotein that delivers genes at levels comparable posttraumatic stress disorder. Arch. Gen. Psychiatry during the consummatory phase of a discriminative
to wild type viral glycoproteins. J.Virol. 86, 373381 50, 295305 (1993). stimulus task depends on previous reward
(2011). 86. Rauch, S.L., Shin, L.M. & Phelps, E.A. Neurocircuitry predictive cues. J.Neurophysiol. 91, 18661882
61. Stuber, G.D. etal. Excitatory transmission from the models of posttraumatic stress disorder and (2004).
amygdala to nucleus accumbens facilitates reward extinction: human neuroimaging researchpast, 111. Carr, G.D., Fibiger, H.C. & Phillips, A.G. Conditioned
seeking. Nature 475, 377380 (2011). present, and future. Biol. Psychiatry 60, 376382 place preference as a measure of drug reward. in The
This study showed that amygdalar projections to (2006). Neuropharmacological Basis of Reward (Topics in
the NAc, but not cortical projections, mediate 87. Milad, M.R., Rauch, S.L., Pitman, R.K. & Quirk, G.J. Experimental Psychopharmacology) Vol. 1 (eds
ICSS. Fear extinction in rats: implications for human brain Liebman, J.M. & Cooper, S.J.) 264319 (Oxford
62. Zorzos, A.N., Boyden, E.S. & Fonstad, C.G. imaging and anxiety disorders. Biol. Psychology 73, Univ. Press, 1989).
Multiwaveguide implantable probe for light delivery 6171 (2006). 112. Tzschentke, T.M. Measuring reward with the
to sets of distributed brain targets. Opt. Lett. 35, 88. Rudy, J.W. & OReilly, R.C. Contextual fear conditioned place preference paradigm: a
41334135 (2010). conditioning, conjunctive representations, pattern comprehensive review of drug effects, recent progress
63. Wentz, C.T. etal. A wirelessly powered and controlled completion, and the hippocampus. Behav. Neurosci. and new issues. Prog. Neurobiol. 56, 613672
device for optical neural control of freely-behaving 113, 867880 (1999). (1998).
animals. J.Neural Eng. 8, 046021 (2011). 89. Fanselow, M.S. Contextual fear, gestalt memories, 113. Lobo, M.K. etal. Cell type-specific loss of BDNF
64. Kessler, R.C. etal. Lifetime prevalence and and the hippocampus. Behav. Brain Res. 110, 7381 signaling mimics optogenetic control of cocaine
ageofonset distributions of DSM-IV disorders in the (2000). reward. Science 330, 385390 (2010).
National Comorbidity Survey Replication. Arch. Gen. 90. Phillips, R. & LeDoux, J. Differential contribution of This study showed that D1 and D2 neurons in the
Psychiatry 62, 593602 (2005). amygdala and hippocampus to cued and contextual NAc have distinctly different effects on cocaine-
65. Lieb, R. Anxiety disorders: clinical presentation and fear conditioning. Behav. Neurosci. 106, 274285 conditioned place preference.
epidemiology. Handb. Exp. Pharmacol. 405432 (1992). 114. Olds, J. & Milner, P. Positive reinforcement produced
(2005). 91. Kim, J.J., Rison, R.A. & Fanselow, M.S. Effects of by electrical stimulation of septal area and other
66. Baldwin, D., Woods, R., Lawson, R. & Taylor, D. amygdala, hippocampus, and periaqueductal gray regions of rat brain. J.Comp. Physiol. Psychol. 47,
Efficacy of drug treatments for generalised anxiety lesions on short-and long-term contextual fear. Behav. 419427 (1954).
disorder: systematic review and meta-analysis. BMJ Neurosci. 107, 10931098 (1993). 115. Lscher, C. & Malenka, R.C. Drug-evoked synaptic
342, d1199 (2011). 92. McGaugh, J.L. Memorya century of consolidation. plasticity in addiction: from molecular changes to
67. Beck, A.T., Emery, G. & Greenberg, R.L. Anxiety Science 287, 248251 (2000). circuit remodeling. Neuron 69, 650663 (2011).
Disorders and Phobias: A Cognitive Perspective (Basic 93. Frankland, P.W. etal. Consolidation of CS and US 116. Schoenbaum, G., Chiba, A.A. & Gallagher, M.
Books, 2005). representations in associative fear conditioning. Orbitofrontal cortex and basolateral amygdala encode
68. Woods, J.H., Katz, J.L. & Winger, G. Benzodiazepines: Hippocampus 14, 557569 (2004). expected outcomes during learning. Nature Neurosci.
use, abuse, and consequences. Pharmacol. Rev. 44, 94. Frankland, P.W. & Bontempi, B. The organization of 1, 155159 (1998).
151347 (1992). recent and remote memories. Nature Rev. 117. Schoenbaum, G. & Roesch, M. Orbitofrontal cortex,
69. Johansen, J.P. etal. Optical activation of lateral Neuroscience 6, 119130 (2005). associative learning, and expectancies. Neuron 47,
amygdala pyramidal cells instructs associative 95. Wiltgen, B.J., Brown, R.A.M., Talton, L.E. & Silva, 633636 (2005).
fear learning. Proc. Natl Acad. Sci. USA 107, A.J. New circuits for old memories: the role of the 118. Tye, K.M., Stuber, G.D., de Ridder, B., Bonci, A. &
1269212697 (2010). neocortex in consolidation. Neuron 44, 101108 Janak, P.H. Rapid strengthening of thalamo-amygdala
70. Adolphs, R., Tranel, D., Damasio, H. & Damasio, A. (2004). synapses mediates cue-reward learning. Nature 453,
Fear and the human amygdala. J.Neurosci. 15, 96. Lee, J.L.C., Everitt, B.J. & Thomas, K.L. Independent 12531257 (2008).
58795891 (1995). cellular processes for hippocampal memory 119. Tye, K.M. etal. Methylphenidate facilitates learning-
71. Davis, M. The role of the amygdala in fear and anxiety. consolidation and reconsolidation. Science 304, induced amygdala plasticity. Nature Neurosci. 13,
Annu. Rev. Neurosci. 15, 353375 (1992). 839843 (2004). 475481 (2010).

264 | APRIL 2012 | VOLUME 13 www.nature.com/reviews/neuro

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

120. Carelli, R.M. & Deadwyler, S. A comparison of nucleus 143. Mervaala, E. etal. Quantitative MRI of the 168. Tnnesen, J., Srensen, A.T., Deisseroth, K.,
accumbens neuronal firing patterns during cocaine hippocampus and amygdala in severe depression. Lundberg, C. & Kokaia, M. Optogenetic control of
self-administration and water reinforcement in rats. Psychol. Med. 30, 117125 (2000). epileptiform activity. Proc. Natl Acad. Sci. USA 106,
J.Neurosci. 14, 77354776 (1994). 144. Sheline, Y.I., Gado, M.H. & Price, J.L. Amygdala core 1216212167 (2009).
121. Carelli, R.M., Ijames, S.G. & Crumling, A.J. Evidence nuclei volumes are decreased in recurrent major 169. Paz, J.T. etal. A new mode of corticothalamic
that separate neural circuits in the nucleus accumbens depression. Neuroreport 9, 20232028 (1998). transmission revealed in the Gria4/ model of
encode cocaine versus natural (water and food) 145. Sheline, Y.I. etal. Increased amygdala response to absence epilepsy. Nature Neurosci. 14, 11671173
reward. J.Neurosci. 20, 42554266 (2000). masked emotional faces in depressed subjects (2011).
122. Wakabayashi, K.T., Fields, H.L. & Nicola, S.M. resolves with antidepressant treatment: an fMRI 170. Kim, J.A., Szatmari, P., Bryson, S.E., Streiner, D.L. &
Dissociation of the role of nucleus accumbens study. Biol. Psychiatry 50, 651658 (2001). Wilson, F.J. The prevalence of anxiety and mood
dopamine in responding to reward-predictive cues and 146. Siegle, G.J., Thompson, W., Carter, C.S., problems among children with autism and Asperger
waiting for reward. Behav. Brain Res. 154, 1930 Steinhauer, S.R. & Thase, M.E. Increased amygdala syndrome. Autism 4, 117132 (2000).
(2004). and decreased dorsolateral prefrontal BOLD 171. Markram, K. & Markram, H. The intense world
123. Peters, Y.M., ODonnell, P. & Carelli, R.M. Prefrontal responses in unipolar depression: related and theorya unifying theory of the neurobiology of
cortical cell firing during maintenance, extinction, and independent features. Biol. Psychiatry 61, 198209 autism. Front. Hum. Neurosci. 4, 224 (2010).
reinstatement of goal-directed behavior for natural (2007). 172. Vattikuti, S. & Chow, C.C. A computational model
reward. Synapse 56, 7483 (2005). 147. Larisch, R. etal. In vivo evidence for the involvement for cerebral cortical dysfunction in autism
124. Tye, K.M., Cone, J.J., Schairer, W.W. & Janak, P.H. of dopamineD2 receptors in striatum and anterior spectrum disorders. Biol. Psychiatry 67, 672678
Amygdala neural encoding of the absence of reward cingulate gyrus in major depression. Neuroimage 5, (2010).
during extinction. J.Neurosci. 30, 116125 (2010). 251260 (1997). 173. Rubenstein, J. & Merzenich, M. Model of autism:
125. Tye, K.M. & Janak, P.H. Amygdala neurons 148. Zhu, X., Peng, S., Zhang, S. & Zhang, X. Stress-induced increased ratio of excitation/inhibition in key neural
differentially encode motivation and reinforcement. depressive behaviors are correlated with Par4 and systems. Genes Brain Behav. 2, 255267 (2003).
J.Neurosci. 27, 39373945 (2007). DRD2 expression in rat striatum. Behav. Brain Res. 174. Orekhova, E.V. etal. Excess of high frequency
126. Phillips, P.E.M., Stuber, G.D., Heien, M., Wightman, 223, 329335 (2011). electroencephalogram oscillations in boys with autism.
R.M. & Carelli, R.M. Subsecond dopamine release 149. Meyer, J.H. etal. Lower dopamine transporter Biol. Psychiatry 62, 10221029 (2007).
promotes cocaine seeking. Nature 422, 614618 binding potential in striatum during depression. 175. Kalivas, P.W. & Volkow, N.D. The neural basis of
(2003). Neuroreport 12, 41214125 (2001). addiction: a pathology of motivation and choice. Am.
127. Alsi, J. etal. Enhanced sucrose and cocaine self- 150. Binfar, R.W., Mantovani, M., Budni, J., Santos, J.Psychiatry 162, 14031413 (2005).
administration and cue-induced drug seeking after A.R.S. & Rodrigues, A.L.S. Involvement of dopamine 176. Batel, P. Addiction and schizophrenia. Eur. Psychiatry
loss of VGLUT2 in midbrain dopamine neurons in mice. receptors in the antidepressant-like effect of melatonin 15, 115122 (2000).
J.Neurosci. 31, 1259312603 (2011). in the tail suspension test. Eur. J.Pharmacol. 638, 177. Huppert, J.D., Weiss, K.A., Lim, R., Pratt, S. & Smith,
128. Blazer, D., Kessler, R., McGonagle, K. & Swartz, M. 7883 (2010). T.E. Quality of life in schizophrenia: contributions of
The prevalence and distribution of major depression in 151. Willner, P. Dopamine and depression: a review of anxiety and depression. Schizophr. Res. 51, 171180
a national community sample: the National recent evidence. II. Theoretical approaches. Brain Res. (2001).
Comorbidity Survey. Am. J.Psychiatry 151, 979986 Rev. 6, 225236 (1983). 178. Rasmussen, K. CCK, schizophrenia, and anxiety. Ann.
(1994). 152. Malone, D.A. Jr etal. Deep brain stimulation of the NY Acad. Sci. 713, 300311 (1994).
129. Nierenberg, A.A. & Dececco, L.M. Definitions of ventral capsule/ventral striatum for treatment- 179. Busskamp, V. etal. Genetic reactivation of cone
antidepressant treatment response, remission, resistant depression. Biol. Psychiatry 65, 267275 photoreceptors restores visual responses in retinitis
nonresponse, partial response, and other relevant (2009). pigmentosa. Science 329, 413417 (2010).
outcomes: a focus on treatment-resistant depression. 153. Aghajanian, G.K., Graham, A.W. & Sheard, M.H. 180. Busskamp, V. & Roska, B. Optogenetic approaches to
J.Clin. Psychiatry 62 (Suppl. 16), 59 (2001). Serotonin-containing neurons in brain: depression of restoring visual function in retinitis pigmentosa. Curr.
130. Dailly, E., Chenu, F., Renard, C.E. & Bourin, M. firing by monoamine oxidase inhibitors. Science 169, Opin. Neurobiol. 21, 942946 (2011).
Dopamine, depression and antidepressants. Fundam. 11001102 (1970). 181. Pagliardini, S. etal. Active expiration induced by
Clin. Pharmacol. 18, 601607 (2004). 154. Arango, V., Underwood, M.D. & Mann, J.J. Serotonin excitation of ventral medulla in adult anesthetized
131. Maddox, J.C., Levi, M. & Thompson, C. The brain circuits involved in major depression and suicide. rats. J.Neurosci. 31, 28952905 (2011).
compliance with antidepressants in general practice. Prog. Brain Res. 136, 443453 (2002). 182. Gourine, A.V. etal. Astrocytes control breathing
J.Psychopharmacol. 8, 4852 (1994). 155. Weiss, J.M. etal. Behavioral depression produced by through pH-dependent release of ATP. Science 329,
132. Melfi, C.A. etal. The effects of adherence to an uncontrollable stressor: relationship to 571575 (2010).
antidepressant treatment guidelines on relapse and norepinephrine, dopamine, and serotonin levels in 183. Alexander, G.E. & Crutcher, M.D. Functional
recurrence of depression. Arch. Gen. Psychiatry 55, various regions of rat brain. Brain Res. Rev. 3, architecture of basal ganglia circuits: neural substrates
11281132 (1998). 167205 (1981). of parallel processing. Trends Neurosci. 13, 266271
133. Mayberg, H.S. etal. Deep brain stimulation for 156. Swerdlow, N.R. & Koob, G.F. Dopamine, (1990).
treatment-resistant depression. Neuron 45, 651660 schizophrenia, mania, and depression: toward a 184. Gurney, K., Prescott, T. & Redgrave, P. A
(2005). unified hypothesis of corticostriatopallido-thalamic computational model of action selection in the basal
A seminal study showing that DBS can show acute function. Behav. Brain Sci. 10, 197245 (1987). ganglia. I. A new functional anatomy. Biol. Cybern. 84,
and long-lasting antidepressant effects, inviting 157. Li, X., Frye, M.A. & Shelton, R.C. Review of 401410 (2001).
reverse translational studies using optogenetics to pharmacological treatment in mood disorders and 185. Smith, Y., Bevan, M., Shink, E. & Bolam, J.
provide a mechanistic explanation. future directions for drug development. Microcircuitry of the direct and indirect pathways of
134. Mayberg, H. Modulating limbic-cortical circuits in Neuropsychopharmacology 37, 77101 (2012). the basal ganglia. Neuroscience 86, 353387
depression: targets of antidepressant treatments. 158. Gillott, A., Furniss, F. & Walter, A. Anxiety in high- (1998).
Semin. Clin. Neuropsychiatry 7, 255268 functioning children with autism. Autism 5, 277286 186. Hughes, A.J., Daniel, S.E., Kilford, L. & Lees, A.J.
(2002). (2001). Accuracy of clinical diagnosis of idiopathic Parkinsons
135. Covington, H.E. etal. Antidepressant effect of 159. Howlin, P. & Moorf, A. Diagnosis in autism. Autism 1, disease: a clinico-pathological study of 100 cases.
optogenetic stimulation of the medial prefrontal 135162 (1997). J.Neurol. Neurosurg. Psychiatry 55, 181184
cortex. J.Neurosci. 30, 1608216090 (2010). 160. Phetrasuwan, S., Miles, M.S., Mesibov, G.B. & (1992).
136. Bach-Mizrachi, H. etal. Neuronal tryptophan Robinson, C. Defining autism spectrum disorders. 187. Gelb, D.J., Oliver, E. & Gilman, S. Diagnostic criteria
hydroxylase mRNA expression in the human dorsal J.Spec. Pediatr. Nurs. 14, 206209 (2009). for parkinson disease. Arch. Neurol. 56, 3339
and median raphe nuclei: major depression and 161. Crow, T.J. Molecular pathology of schizophrenia: more (1999).
suicide. Neuropsychopharmacology 31, 814824 than one disease process? Br. Med. J. 280, 6668 188. Benabid, A.L. Deep brain stimulation for Parkinsons
(2005). (1980). disease. Curr. Opin. Neurobiol. 13, 696706 (2003).
137. Greenwood, B.N. etal. Freewheel running prevents 162. Endicott, J. & Spitzer, R.L. A diagnostic interview: the 189. Deuschl, G. etal. A randomized trial of deep-brain
learned helplessness/behavioral depression: role of schedule for affective disorders and schizophrenia. stimulation for Parkinsons disease. N.Engl. J.Med.
dorsal raphe serotonergic neurons. J.Neurosci. 23, Arch. Gen. Psychiatry 35, 837844 (1978). 355, 896908 (2006).
28892898 (2003). 163. Kay, S.R., Flszbein, A. & Opfer, L.A. The positive and 190. Kumar, R. etal. Double-blind evaluation of
138. Lira, A. etal. Altered depression-related behaviors negative syndrome scale (PANSS) for schizophrenia. subthalamic nucleus deep brain stimulation in
and functional changes in the dorsal raphe nucleus of Schizophr. Bull. 13, 261276 (1987). advanced Parkinsons disease. Neurology 51,
serotonin transporter-deficient mice. Biol. Psychiatry 164. Kehrer, C., Maziashvili, N., Dugladze, T. & Gloveli, T. 850855 (1998).
54, 960971 (2003). Altered excitatory-inhibitory balance in the NMDA- 191. Byers, B. etal. SNCA triplication Parkinsons patients
139. Airan, R.D. etal. High-speed imaging reveals hypofunction model of schizophrenia. Front. Mol. iPSC-derived DA neurons accumulate synuclein and
neurophysiological links to behavior in an animal Neurosci. 1, 6 (2008). are susceptible to oxidative stress. PLoS ONE 6,
model of depression. Science 317, 819823 165. Gonzalez-Burgos, G., Hashimoto, T. & Lewis, D.A. e26159 (2011).
(2007). Alterations of cortical GABA neurons and network 192. Lin, D. etal. Functional identification of an aggression
140. Bremner, J.D. etal. Hippocampal volume reduction in oscillations in schizophrenia. Curr. Psychiatry Rep. 12, locus in the mouse hypothalamus. Nature 470,
major depression. Am. J.Psychiatry 157, 115118 335344 (2010). 221226 (2011).
(2000). 166. Lewis, D.A. & Gonzalez-Burgos, G. This study used optogenetic manipulation of
141. Sahay, A. & Hen, R. Adult hippocampal neurogenesis Pathophysiologically based treatment interventions in neurons to show the causal role of this
in depression. Nature Neurosci. 10, 11101115 schizophrenia. Nature Med. 12, 10161022 (2006). subpopulation of neurons in mediating aggressive
(2007). 167. Lord, C. & Bishop, S.L. Social Policy Report 24. behaviours.
142. Sapolsky, R.M. The possibility of neurotoxicity in the Autism Spectrum Disorders: Diagnosis, Prevalence, 193. Lin, S.C., Deisseroth, K. & Henderson, J.M.
hippocampus in major depression: a primer on neuron and Services for Children and Families (Society for Optogenetics: background and concepts for
death. Biol. Psychiatry 48, 755765 (2000). Research in Child Development, 2010). neurosurgery. Neurosurgery 69, 13 (2011).

NATURE REVIEWS | NEUROSCIENCE VOLUME 13 | APRIL 2012 | 265

2012 Macmillan Publishers Limited. All rights reserved


REVIEWS

194. Han, X. etal. Millisecond-timescale optical Acknowledgements founders of a company (Circuit Therapeutics) focused on drug
control of neural dynamics in the nonhuman We would like to acknowledge E. E. Steinberg, P. H. Janak, screening and optogenetic therapies for bladder dysfunction.
primate brain. Neuron 62, 191198 S.Y. Kim, L. D. Tye, O. Yizhar, A. Kreitzer, A. Kravitz and L. E. K.D. and K.M.T. have no more than 5% interest in any com-
(2009). Fenno for helpful comments and discussion. We thank the pany and receive no research funding, royalties or consultant
195. Berdyyeva, T.K. & Reynolds, J.H. The dawning of Deisseroth laboratory for intellectual and scientific support. fees from any company. All optogenetic tools and methods
primate optogenetics. Neuron 62, 159160 K.M.T. is supported by the US National Institute of Mental are distributed and supported freely from the laboratory
(2009). Health (NIMH) (1F32MH08801001), the Picower Institute (http://www.optogenetics.org).
196. Lewis, T.L., Mao, T., Svoboda, K. & Arnold, D.B. of Learning and Memory and the Picower Institute Innovation
Myosin-dependent targeting of transmembrane Funds (PIIF). K.D. is supported by the NIMH, the US National Competing interests statement
proteins to neuronal dendrites. Nature Neurosci. 12, Institute on Drug Abuse, the US National Institute of The authors declare no competing financial interests.
568576 (2009). Neurological Disorders and Stroke, the Howard Hughes
197. Kleinlogel, S. etal. Ultra light-sensitive and fast Medical Institute, the Defense Advanced Research Projects
neuronal activation with the Ca2+-permeable Agency Reorganization and Plasticity to Accelerate Injury FURTHER INFORMATION
channelrhodopsin CatCh. Nature Neurosci. 14, Recovery Program, the Keck Foundation, the Wiegers Family Kay M. Tyes homepage: http://www.tyelab.org
513518 (2011). Gift Fund and the Gatsby Charitable Foundation. Full funding Karl Deisseroths homepage: http://www.stanford.edu/
198. Kato, H. E. et al. Crystal structure of the support is described at http://www.optogenetics.org/funding. group/dlab/
channelrhodopsin light-gated cation channel. Nature Stanford University has filed for patent protection on tech- ALL LINKS ARE ACTIVE IN THE ONLINE PDF
482, 369374 (2012). nologies invented by K.M.T. and K.D., and K.D. is one of the

266 | APRIL 2012 | VOLUME 13 www.nature.com/reviews/neuro

2012 Macmillan Publishers Limited. All rights reserved

You might also like