Sie sind auf Seite 1von 19

LETTER doi:10.

1038/nature21047

A competitive inhibitory circuit for selection of


active and passive fear responses
Jonathan P. Fadok1, Sabine Krabbe1, Milica Markovic1,2, Julien Courtin1, Chun Xu1, Lema Massi1, Paolo Botta1,2†, Kristine Bylund1,
Christian Müller1, Aleksandar Kovacevic1, Philip Tovote1 & Andreas Lüthi1,2

When faced with threat, the survival of an organism is contingent conditioned flight response was characterized by increases in speed
upon the selection of appropriate active or passive behavioural (Fig. 1d) and the number of escape jumps (Fig. 1d, Supplementary
responses1–3. Freezing is an evolutionarily conserved passive fear Video 1). This behaviour was quantified as a flight score (Fig. 1c, d, see
response that has been used extensively to study the neuronal Methods). Behavioural analysis revealed that tone exposure signifi-
mechanisms of fear and fear conditioning in rodents4. However, cantly enhanced freezing compared to contextual freezing, and that
rodents also exhibit active responses such as flight under natural exposure to white noise, concomitant with the observed increase in
conditions2. The central amygdala (CEA) is a forebrain structure flight score, resulted in a significant reduction of the time spent freezing
vital for the acquisition and expression of conditioned fear (Fig. 1d). Importantly, the white noise stimulus was not in itself aversive
responses, and the role of specific neuronal sub-populations of (Extended Data Fig. 1).
the CEA in freezing behaviour is well-established1,5–7. Whether To test for the effect of threat proximity and context2,8 on conditioned
the CEA is also involved in flight behaviour, and how neuronal flight, mice were split into two groups: one that underwent extinction
circuits for active and passive fear behaviour interact within the training in the conditioning context, and one that was exposed to the
CEA, are not yet understood. Here, using in vivo optogenetics and SCS in a neutral context (Fig. 1a, e, f). Consistent with threat immi-
extracellular recordings of identified cell types in a behavioural nence theory8, conditioned flight behaviour was rapidly extinguished
model in which mice switch between conditioned freezing and upon removal of the footshock during extinction (Fig. 1e), and was also
flight, we show that active and passive fear responses are mediated context-dependent (Fig. 1e, f). In line with the hierarchical organiza-
by distinct and mutually inhibitory CEA neurons. Cells expressing tion of distinct defensive behaviours2, extinction of conditioned flight
corticotropin-releasing factor (CRF+) mediate conditioned flight, behaviour was accompanied by an increase in freezing in response to
and activation of somatostatin-positive (SOM+) neurons initiates the white noise (Fig. 1e).
passive freezing behaviour. Moreover, we find that the balance The CEA is a striatum-like structure comprised of distinct neuronal
between conditioned flight and freezing behaviour is regulated by populations of inhibitory GABAergic neurons that express CRF, SOM
means of local inhibitory connections between CRF+ and SOM+ or protein kinase C δ​ (PKCδ​)11–14, among other molecular markers15
neurons, indicating that the selection of appropriate behavioural (see also Extended Data Fig. 2). The CEA can be subdivided into the
responses to threat is based on competitive interactions between two lateral CEA (CEl) and medial CEA (CEm). Disinhibition of CEm
defined populations of inhibitory neurons, a circuit motif allowing output neurons by inhibitory interactions between CEl SOM+ and
for rapid and flexible action selection. PKCδ​+ neurons gates the expression of conditioned freezing11,14,16,17.
Mammals express adaptive behavioural responses to threat1,2. The Furthermore, suppression of activity within CEA cellular substrates of
type of response depends on intensity and proximity of threat, as freezing leads to active behavioural coping with fear18. To d ­ etermine
well as context2,8. Although different behavioural fear responses are the role of defined CEA neuronal subpopulations for expression of
inborn, associative learning enables organisms to use environmental conditioned flight, we used the inhibitory opsin a­ rchaerhodopsin
cues to predict and respond to danger9. Pavlovian fear conditioning is (Arch), which was Cre-dependently expressed in CRF+, SOM+
one ­paradigm that has been used to understand the neural circuitry and PKCδ​+ neurons (Fig. 2a and Extended Data Fig. 2). Efficacy of
underlying associative learning5,9,10. In Pavlovian fear conditioning, a Arch-mediated neuronal inhibition was tested using extracellular
conditioned stimulus (CS; for example, an auditory stimulus) is paired recordings of identified units in behaving animals (Extended Data
with an aversive unconditioned stimulus (US; for example, electrical Fig. 2, see also Methods). Inhibition of CRF+, but not SOM+ or
footshock). Electrical footshock, the most commonly used US, elicits a PKCδ​+ neurons, completely abolished conditioned flight behaviour
variety of defensive responses, including flight and freezing4; ­however, (Fig. 2b–g and Extended Data Fig. 2). Optogenetic inhibition in naive
freezing is the dominant conditioned response to the CS in most mice did not elicit any freezing or flight behaviour (Extended Data Fig. 2).
fear-conditioning experiments5. Fluorophore-expressing somata were predominately found in CEl
We developed a Pavlovian conditioning paradigm that promotes (89 ±​ 3% CRF–Arch, 91 ±​ 4% SOM–Arch, and 100% PKCδ​–Arch11);
active defensive behaviour, including flight. Conditioned flight however, minor expression was also observed in the CEm. Together,
responses to the CS were elicited by extending standard conditioning these data demonstrate that CEA CRF+ neurons are necessary for the
protocols (Extended Data Fig. 1). To allow for direct, within-subject expression of conditioned flight.
comparison of active and passive behaviours, we tested different CSs We next determined the electrophysiological response profile of
and found that a serial compound stimulus (SCS) consisting of a pure CRF+ neurons during conditioned flight. Channelrhodopsin-2 (ChR2)
tone followed by white noise induced rapid behavioural switches or Arch was expressed in CEA CRF+ neurons, and mice were subse-
between conditioned freezing and flight behaviour (Fig. 1a–d). With quently implanted with optrodes (Fig. 3a and Extended Data Fig. 3).
conditioning, mice exhibited freezing in response to the tone, and Single units were recorded and isolated using standard procedures16
flight during the white noise (Fig. 1c, d, Supplementary Video 1). The and CRF+ cells were identified by short-latency responses to light

1
Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland. 2University of Basel, 4000 Basel, Switzerland. †Present address: Champalimaud Centre for the
Unknown, Av. Brasilia, 1400-038 Lisbon, Portugal.

0 0 M O N T H 2 0 1 7 | VO L 0 0 0 | NAT U R E | 1
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

a Behavioural paradigm Context A


Day 4
b Stimuli (Fig. 3b, Extended Data Fig. 3 and Methods). CRF+ cells exhibited an
Context B
Extinction SCS US increase in responses to white noise between days 1 and 3 (Fig. 3c, d and
Day 1 Day 2 Day 3
16 × SCS
Extended Data Fig. 3); however, in the neutral context, where flight was
4 × SCS 5 × SCS–US 5 × SCS–US
4 × SCS
never observed, CRF+ excitation in response to white noise was dimi­
Pre-exposure Conditioning nished compared to day 3 (Fig. 3e). By normalizing firing rates during
Retrieval Tone White noise
epochs of conditioned flight and freezing behaviour, more than half
c Pre-
exposure Conditioning
d Day 3, conditioning of the CRF+ cells (5 of 8) were found to be excited specifically during
Day 1 Day 2 Day 3
20 flight, but not during freezing behaviour (Fig. 3i and Methods). These
Speed (cm s–1) Pre-CS
18 15
Tone
findings are consistent with the notion that activity of CRF+ neurons
Flight score (AU)

14
10 White noise contributes to conditioned flight behaviour.
10
5 Shock
Previous studies suggested that activation of CEl SOM+ cells is
6
0
0 10 20 30 40
important for conditioned freezing14. We therefore recorded from
2 Time (s) identified CEl SOM+ neurons during the conditioned flight ­paradigm
Pre-CS Tone White noise Shock (Fig. 3a and Extended Data Fig. 3). Before conditioning, there was
Tone
White noise
*** no significant response of identified SOM+ cells to either the pure
100 20
***
20 100 *** ***
*** tone or to white noise (Fig. 3f and Extended Data Fig. 3). Following
Number of jumps

Flight score (AU)

80 80
Freezing (%)

15 15
­conditioning, SOM+ cells became strongly inhibited by white noise
Freezing (%)
60 * 60
40
10 10 (Fig. 3g). However, they exhibited significantly elevated firing rates
40
20 5 5
during the 3 min baseline, a period of high levels of contextual ­freezing
20
0
(pre-­exposure firing rate: 2.7 ±​ 0.6 spikes per s; day 3 firing rate:
Trial 1 2 3 4 1 3 5 1 3 5
0 0 0
5.8 ±​ 1.3 spikes per s; two-tailed paired t-test, P <​  0.05). Consistent
e Day 4, extinction f Day 4, retrieval
with this, SOM+ neurons were excited by both conditioned stimuli
8 20 *** 8 8 in the neutral context, in which mice displayed freezing behaviour in
Flight score (AU)
Flight score (AU)

Flight score (AU)

6 15 6 6 response to both pure tone and white noise (Fig. 3h and Extended Data
4 10 4 4 Fig. 3). When analysing the activity of SOM+ neurons as a function
2 5 2 2 ** of behaviour, we found that the majority of SOM+ neurons (6 of 7)
0 0 0 0 were activated during freezing, but inhibited during flight, pointing
Tone
*** *** towards an interaction between freezing and flight pathways (Fig. 3i
100 White noise 100 100 100
and Methods).
80 80 80 80
To address this, we performed optogenetic gain-of-function experi­
Freezing (%)

Freezing (%)

Freezing (%)

60 60 60 60
ments (Fig. 4a and Extended Data Fig. 4). Photo-excitation of ChR2-
40 40 40 40
expressing CRF+ neurons during tone exposure decreased freezing and
20 20 20 20
led to more active behaviour in the conditioning context (Fig. 4b–d),
0 0 0
Trial 1 4 8 12 16 Trial 1–4 Trial 1 2 3 4
0
Trial 1–4 and decreased freezing in response to the SCS in the neutral context
(Extended Data Fig. 4). There was no effect on freezing or flight in naive
Figure 1 | Conditioned flight paradigm. a, Protocol used to elicit animals (Extended Data Fig. 4). Therefore, in conditioned mice, exo­
conditioned flight responses. On day 4, mice were split into two groups. genous excitation of CRF+ neurons is sufficient to bias fear responses
b, The conditioned stimulus is a serial compound stimulus (SCS) which
away from passive freezing behaviour and towards more active defen-
is paired with a 1 s footshock during conditioning. c, Top, comparison of
flight scores across sessions (day 1 to day 3, n =​ 20, two-way repeated-
sive behaviour.
measures ANOVA, cue ×​trial interaction, F(13,247) =​  4.82, P <​  0.01). Conversely, optical excitation of SOM+ neurons during exposure to
AU, arbitrary units. Bottom, comparison of freezing responses across white noise decreased flight and caused an increase in freezing behaviour
sessions (day 1 to day 3, n =​ 20; two-way repeated-measures ANOVA, (Fig. 4b–d), in line with the observed neuronal activation patterns (Fig. 3),
cue ×​trial interaction, F(13,247) =​  18.3, P <​  0.01). d, Top, average speed and a recent finding suggesting a role for this neuronal subtype in
trace of mice (n =​ 10) from day 3, note increase in speed during exposure modu­lating behavioural output19. Consistent with previous reports14,
to white noise. Bottom left, the number of jump escape responses on day we also found that ChR2-mediated stimulation of SOM+ cells induced
3 (n =​ 20, Friedman test, P <​ 0.01, Dunn’s multiple comparisons test). unconditioned freezing in naive SOM–ChR2 mice (Extended Data
Bottom centre, flight scores on day 3 (n =​ 20, Wilcoxon matched-pairs Fig. 4).
signed-rank test). Bottom right, freezing behaviour on day 3 (n =​  20;
One possible mechanism that could account for the mutual exclusion
one-way repeated-measures ANOVA, F =​  56.82, P <​ 0.01, Tukey’s multiple
comparisons test). e, Top left, rapid extinction of conditioned flight between the freezing and the flight behaviours could be a reciprocal
(n =​ 12, two-way repeated-measures ANOVA, cue ×​trial interaction, inhibitory connection between CRF+ and SOM+ neurons within the
F(15,165) =​  3.05, P <​ 0.01). Top right, flight scores from first block of local CEA network. To test this, we expressed ChR2–eYFP in CEA
four trials of extinction (n =​ 12, Wilcoxon matched-pairs signed-rank CRF+ cells and recorded inhibitory postsynaptic currents (IPSCs) in
test). Bottom left, freezing in response to white noise increases during non-labelled CEl target neurons of acute brain slices during light stimu-
extinction (n =​ 12, two-way repeated-measures ANOVA, cue ×​ trial lation of the CRF+ network (Fig. 4e–h and Extended Data Fig. 5). While
interaction, F(15,165) =​  3.55, P <​ 0.01). Bottom right, freezing for the first both SOM+ and PKCδ​+ neurons receive GABAergic inhibitory input
block of four trials during extinction (n =​ 12, Wilcoxon matched-pairs from CRF+ cells (Fig. 4f, h and Extended Data Fig. 5), we observed
signed-rank test). f, Top left, conditioned flight is context-dependent
that there were significantly more SOM+ than PKCδ​+ cells receiving
(n =​ 8, two-way repeated-measures ANOVA, cue ×​trial interaction,
F(1,7) =​  27.44, P <​ 0.01). Top right, flight scores in the neutral context
inhibition from CRF+ neurons (Fig. 4g). These data suggest that the
(two-tailed paired t-test). Bottom left, freezing responses across trials CRF+ network, which is vital for conditioned flight, provides direct
during retrieval (n =​ 8, two-way repeated-measures ANOVA, effect of inhibition to the SOM+ network, which encodes freezing behaviour.
cue F(1,7) =​  27.67, P <​ 0.01). Bottom right, freezing responses during We next optically activated SOM+ neurons and recorded from
retrieval are highest in response to white noise (n =​ 8, two-tailed paired identified CRF+ and non-labelled target neurons in the CEl (Fig. 4i–l
t-test). Box and whisker plots indicate median, interquartile range, and Extended Data Fig. 5). These experiments revealed predominant
and 5th to 95th percentiles of the distribution, crosses indicate GABAergic inhibitory connections from SOM+ onto CRF+ neurons
means. All other values are means ±​  s.e.m. *​P <​  0.05; *​*​P <​  0.01; (Fig. 4j–l and Extended Data Fig. 5). Importantly, evoked spiking
*​*​*​P <​  0.001. activity of both CRF+ and SOM+ neurons could be blocked by

2 | NAT U R E | VO L 0 0 0 | 0 0 M O N T H 2 0 1 7
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

a AAV–flex–Arch–GFP or b Behavioural paradigm


a AAV–flex–Arch–GFP or b
AAV–flex–GFP
AAV flex GFP Fibre AAV–flex–ChR2–eYFP Fibre Electrode 3
Day 1 Day 2 Day 3

Jitter (ms)
connector connector connector
2
4 × SCS 5 × SCS–US 8 × SCS–US
Spont. 1
Optrode 50 μV Light evok.
Pre-exposure Conditioning 250 μs 0

Waveform similarity (r)


Light CRF SOM
Day 3 10 s 10 s
1.000

Frequency (Hz)
4 × SCS
60
0.975
BLA Mouse lines: Light on BLA 30
CEA CRF-ires-cre CEA Mouse lines:
4 × SCS 0 0.950
SOM-ires-cre White noise CRF-ires-cre 0 0.3
White noise + light SOM-ires-cre Time (s)
c d Day 3 onset g Day 3 white noise
c d e

Change in flight score (%)


8 100 Pre-exposure (day 1) Conditioning (day 3) Retrieval (day 4)
CRF–Arch ** White noise White noise White noise
CEl White noise
Speed (cm s–1)

6 White noise 50 **
+ light
4 0
BLA
2 –50 4
CEm CRF+
3 cells

z score
0 –100 2
GFP control
e f Day 3 h CRF–Arch
1
8 100
Change in freezing (%)

SOM–Arch SOM–Arch 0
CEl
White noise –1
Speed (cm s–1)

6 50
White noise
+ light f g h
4 0
BLA
CEm 2 –50
4 SOM+
0 –100
cells

z score
–10
10 –5
5 0 5 10 2
Time (s)
0
Figure 2 | CRF+ neurons mediate conditioned flight. a, Injection and –2
implantation strategy for loss-of-function experiments. Mouse brain figure –250 0 250 500 –250 0 250 500 –250 0 250 500
reproduced with permission from ref. 31. BLA, basolateral amygdala. Time (ms) Time (ms) Time (ms)
b, Behavioural protocol design for loss-of-function experiments.
i Flight Freezing Flight Freezing
c, Expression of Arch in CRF+ neurons. Bregma −​1.58 mm caudal. Scale 4
bar, 250 μ​m. d, Speed traces of the CRF–Arch group (n =​  7). e, Expression 2
of Arch in SOM+ neurons. Bregma −​1.58 mm caudal. Scale bar, 250 μ​m.
z score

f, Speed traces of the SOM–Arch group (n =​  7). g, Flight scores are lowered 0
by optical inhibition of CRF+, but not SOM+ cells. (GFP control, n =​  6; –2
CRF, n =​  7; SOM, n =​ 7; one-way ANOVA, F =​  9.52, P <​  0.01. Tukey’s CRF+ cells SOM+ cells
multiple comparisons test). h, Change in freezing induced by optical 0 5 10 15 0 5 10 15 0 5 10 15 0 5 10 15
inhibition of CRF+ or SOM+ cells (one-way ANOVA; no significant Time bins Time bins Time bins Time bins
differences). Box and whisker plots indicate median, interquartile range, Figure 3 | Neuronal activity patterns of CRF+ and SOM+ cells.
and 5th to 95th percentiles of the distribution, crosses indicate means. All a, Injection and implantation strategy for identified single-unit recordings.
other values are means ±​  s.e.m. *​*​P <​  0.01. Mouse brain figure reproduced with permission from ref. 31. b, Top left,
example spike waveforms from an identified CRF+ neuron. Bottom left,
excitation of the other population (Fig. 4m–o). Therefore, the cellular example CRF+ unit identified with ChR2. Top right, jitter of light-evoked
substrates of conditioned flight and freezing behaviour are poised to responses. Bottom right, waveform similarity of action potentials in and
inhibit each other through reciprocal inhibitory synaptic connections, out of light. c–h, Normalized activity (z score, bottom) and example raster
a circuit motif which would allow for rapid switching between behav- plots (top) of CEl CRF+ or SOM+ neurons. c, CRF+ neuronal responses
(n =​ 8, 7 mice, from 60 total units) to white noise during pre-exposure
ioural strategies20.
(day 1). d, CRF+ neuronal responses to white noise (n =​ 8) are enhanced
Recent data indicate that the CEA may act as a hierarchy generator on day 3 of the flight paradigm. e, CRF+ excitation to white noise is
between innate and learned fear responses through integration of sen- diminished (n =​ 7) in a neutral context. f, SOM+ neuronal responses
sory information21. Our results identify a novel competitive mechanism (n =​ 10, 7 mice, from 46 total units) to white noise during pre-exposure
in which shifting the balance between two classes of inhibitory neurons (day 1). g, SOM+ neurons are inhibited during white noise on day 3
within the CEA alters learned fear states. Our data demonstrate that (n =​  10). h, SOM+ cells (n =​ 8) are excited by white noise in the neutral
this process is crucial for action selection under stimulus-driven high- context. i, Normalized neuronal activity of identified units during
fear states; other CEA circuit mechanisms may account for expres- behavioural events. CRF+ cells are excited during flight (left, 1.9 s average
sion of defensive behaviours in response to ambiguous threat18,21 duration of behaviour) but not during freezing (left centre, 14.2 s average
duration of behaviour). SOM+ cells are inhibited during flight (right
(see Supplementary Discussion).
centre, 2.0 s average duration of behaviour) and excited during freezing
Monosynaptic rabies tracing revealed putative sources of excita- (right, 7.6 s average duration of behaviour). Horizontal lines in b indicate
tory input to CRF+ and SOM+ cells (Extended Data Fig. 6), which the mean. All other values are means ±​  s.e.m.
could convey information about context and threat level17,22. Increased
CRF+ cellular activity in response to these inputs then locally inhibits CEA cell types to influence action selection. Importantly, expression
SOM+ cells to reduce conditioned freezing. Conversely, excitation of of flight or freezing not only requires diminished neuronal activ-
SOM+ cells acts locally to dampen CRF+ cellular activity, and would ity of SOM+ or CRF+ cells, respectively, but also needs adequate
generate freezing responses via direct or indirect projections to the stimulus or context-driven input to these CEA neuronal subclasses.
periaqueductal grey14,16,17,23. Notably, our data suggests that CRF+ In addition, integration of the signals necessary to orchestrate flight
and SOM+ cells receive differential inputs from presynaptic regions is also probably carried out in the hypothalamus1 and periaqueductal
such as the basolateral amygdala and ventral hippocampus (Extended grey1,23,27,28, regions targeted by CRF+ neurons (Extended Data
Data Fig. 6a–e). Recent studies have demonstrated heterogeneity in Fig. 6f–k). Ultimately, integration of information related to threat is
neuronal coding and behavioural function amongst projections from probably carried out through both local and long-range interactions
these regions24–26, which suggests that they may impinge on specific at multiple sites.

0 0 M O N T H 2 0 1 7 | VO L 0 0 0 | NAT U R E | 3
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

a AAV–flex–ChR2–eYFP b Conditioning (day 3) Figure 4 | CRF+ and SOM+ neurons form a reciprocal inhibitory circuit
or AAV–flex–GFP Fibre
CRF–ChR2 or SOM–ChR2
controlling defensive behaviour. a, Injection and implantation scheme
connector
4 × SCS
10 s 10 s for gain-of-function experiments. Mouse brain figure reproduced with
No light permission from ref. 31. b, Internally controlled behavioural protocol
design. c, Left, speed traces of the CRF–ChR2 group (n =​ 7) during light
CRF–ChR2 Light on
4 × SCS
and no-light trials. Right, speed traces from the SOM–ChR2 group (n =​  8)
Tone + light during light and no-light trials. d, Top, change in flight scores on day 3
CEA
BLA
Mouse lines: SOM–ChR2 Light on (CRF–ChR2, n =​ 7; CRF control, n =​  10; unpaired t-test; SOM–ChR2, n =​  8;
CRF-ires-cre 4 × SCS SOM–GFP control n =​  7; unpaired t-test). Bottom, change in freezing on day
SOM-ires-cre White noise
+ light
3 (CRF–ChR2 versus CRF–GFP control, unpaired t-test test; SOM–ChR2
versus SOM–GFP control, unpaired t-test). e, Whole-cell recordings from
c Tone White noise d Tone White

Change in Change in flight


onset onset 150 *** noise CEl target cells were performed while stimulating ChR2-expressing CRF+

score(%)
3 6 100
Tone White noise 50 ***
cells with blue light. f, Left, ChR2-expressing fibres from CRF+ cells. Right,
Speed (cm s–1)

Tone + light White noise 0 biocytin-filled SOM+ neuron identified with immunohistochemistry.
2 4 + light –50 Bottom, IPSCs from an example SOM+ neuron, evoked by photostimulation
40 * ** of the CRF+ network (individual traces in grey, corresponding average

freezing (%)
1 2
0 trace in black). g, Fraction of neurons receiving inhibitory input upon
0 0
–40
CRF
photostimulation of CRF+ cells (χ2 test, P <​ 0.01; Fisher’s exact test).
–10 –5 0 5 10 –10 –5 0 5 10 –80 SOM h, Average IPSC amplitudes from all recorded neurons (SOM+, n =​  23;
Timee (s) Time (s) Control
PKCδ​+, n =​  8; unidentified, n =​ 13; Kruskal–Wallis test, not significant).
e f g h i, Whole-cell recordings from CEl target neurons were performed while
ChR2 Biocytin SOM
SOM PKCδ stimulating ChR2-expressing SOM+ cells with blue light. j, Left, ChR2-
distribution (%))

100 2,000 Unident.


expressing fibres from SOM+ cells. Right, biocytin-filled CRF+ neuron
IPSC amplitude (pA)

CRF
CRF
ChR
hR2
hR
R2
2
ChR2
** **
SOM
or
20 μm 75 600 identified by tdTomato expression. Bottom, IPSCs from one CRF+ neuron,
PKCδ Light 50 400 evoked by photostimulation of the SOM+ network (individual traces in grey,
on
corresponding average trace in black). k, Fraction of CEA neurons receiving
Relative d

25 200
inhibitory inputs upon photostimulation of SOM+ cells, demonstrating
23/32

Whole-cell
13/17
200 pA

8/28

patch-clamp
patch-clamp
t h l
recording
rec
cording and
an 10 ms 0 0 preferential connectivity of SOM+ onto CRF+ neurons (χ2 test, P <​  0.01;
2

light stimulation
Fisher’s exact test). l, Average IPSC amplitudes from all recorded cells
i j ChR2
2 Biocytin k l CRF (CRF+, n =​  24; PKCδ​+, n =​  7; unidentified, n =​ 8; Kruskal–Wallis test, not
CRF PKCδ significant). m, Example traces from a SOM+ cell, demonstrating spike
(%)
%)

100 800
on (%

Unident.
mplitude (pA)

SOM
S OM *** suppression with ChR2-mediated excitation of the CRF+ network.
distribution
istributio

ChR2
ChR
Ch
hR2
hRR2
2 75 300
20 μm
n, Example traces from a CRF+ cell, demonstrating spike suppression with
IPSC amplitude

CRF
or
PKCδ
Lig
gh
Light
on
on 50 200 ChR2-mediated excitation of the SOM+ network. o, Left, spike probability
am

of SOM+ neurons is significantly reduced with concomitant CRF+ network


Relative d

25 10
100
24/31

excitation (two-tailed paired t-test). Right, CRF+ neuronal activity is


50 pA

7/28
8/17

10
0 mss 0 0 significantly diminished by concomitant SOM+ network activation
(Wilcoxon matched-pairs signed-rank test). Horizontal lines indicate means.
m CRF to SOM n SOM to CRF o
*** *** Box and whisker plots of in vitro experiments indicate median, interquartile
probability (%)

100
range, and 10th to 90th percentiles of the distribution. Crosses indicate
Spike

50 means. Box and whisker plots of behavioural experiments indicate median,


interquartile range, and 5th to 95th percentiles of the distribution. Crosses
20 mV

0
indicate means. All other values are means ±​  s.e.m. *​P <​  0.05; *​*​P <​  0.01;
C ent

C ent

ht t
h t

lig n
lig en

1s
*​*​*​P <​  0.001.
+ urre
C t
r

r
+ urr
ur

ur

Current Current Light


C

+ light

Survival in a volatile world is dependent on rapid selection of appro- 5. Tovote, P., Fadok, J. P. & Lüthi, A. Neuronal circuits for fear and anxiety. Nat. Rev.
priate action programs. Therefore, how discrete neuronal circuit Neurosci. 16, 317–331 (2015).
6. Davis, M. in The Amygdala (ed. J. P. Aggleton) Ch. 6, 213–288 (Oxford
elements interface to generate switches between behavioural states is of University Press, 2000).
prime importance. Previous experimental and theoretical work on the 7. Tsetsenis, T., Ma, X. H., Lo Iacono, L., Beck, S. G. & Gross, C. Suppression of
mechanisms of action selection in striatal circuits suggests an important conditioning to ambiguous cues by pharmacogenetic inhibition of the dentate
gyrus. Nat. Neurosci. 10, 896–902 (2007).
role for local recurrent inhibitory interactions20. Our study indicates 8. Perusini, J. N. & Fanselow, M. S. Neurobehavioral perspectives on the
that this circuit motif may represent a particularly effective and widely distinction between fear and anxiety. Learn. Mem. 22, 417–425 (2015).
used mechanism allowing for the rapid and flexible selection of beha­ 9. Maren, S. Neurobiology of Pavlovian fear conditioning. Annu. Rev. Neurosci. 24,
897–931 (2001).
vioural programs necessary for survival, and suggests that alterations in 10. LeDoux, J. E. Emotion circuits in the brain. Annu. Rev. Neurosci. 23, 155–184
the set points of such circuit switches could contribute to maladaptive (2000).
behaviour in anxiety disorders29,30. 11. Haubensak, W. et al. Genetic dissection of an amygdala microcircuit that gates
conditioned fear. Nature 468, 270–276 (2010).
Online Content Methods, along with any additional Extended Data display items and 12. Roberts, G. W., Woodhams, P. L., Polak, J. M. & Crow, T. J. Distribution of
Source Data, are available in the online version of the paper; references unique to neuropeptides in the limbic system of the rat: the amygdaloid complex.
these sections appear only in the online paper. Neuroscience 7, 99–131 (1982).
13. Swanson, L. W., Sawchenko, P. E., Rivier, J. & Vale, W. W. Organization of ovine
Received 30 March; accepted 5 December 2016. corticotropin-releasing factor immunoreactive cells and fibers in the rat brain:
an immunohistochemical study. Neuroendocrinology 36, 165–186 (1983).
Published online 25 January 2017.
14. Li, H. et al. Experience-dependent modification of a central amygdala fear
circuit. Nat. Neurosci. 16, 332–339 (2013).
1. Gross, C. T. & Canteras, N. S. The many paths to fear. Nat. Rev. Neurosci. 13, 15. Viviani, D. et al. Oxytocin selectively gates fear responses through distinct
651–658 (2012). outputs from the central amygdala. Science 333, 104–107 (2011).
2. Blanchard, D. C. & Blanchard, R. J. in Handbook of Anxiety and Fear Vol. 17 16. Ciocchi, S. et al. Encoding of conditioned fear in central amygdala inhibitory
Handbook of Behavioral Neuroscience (eds R. J. Blanchard, D. C. Blanchard, G. circuits. Nature 468, 277–282 (2010).
Griebel & D. Nutt) Ch. 2.4, 63–79 (Academic Press, 2008). 17. Penzo, M. A., Robert, V. & Li, B. Fear conditioning potentiates synaptic
3. Anderson, D. J. & Adolphs, R. A framework for studying emotions across transmission onto long-range projection neurons in the lateral subdivision of
species. Cell 157, 187–200 (2014). central amygdala. J. Neurosci. 34, 2432–2437 (2014).
4. Fanselow, M. S. Conditioned and unconditional components of post-shock 18. Gozzi, A. et al. A neural switch for active and passive fear. Neuron 67, 656–666
freezing. Pavlov. J. Biol. Sci. 15, 177–182 (1980). (2010).

4 | NAT U R E | VO L 0 0 0 | 0 0 M O N T H 2 0 1 7
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

19. Yu, K., Garcia da Silva, P., Albeanu, D. F. & Li, B. Central amygdala somatostatin Supplementary Information is available in the online version of the paper.
neurons gate passive and active defensive behaviors. J. Neurosci. 36,
6488–6496 (2016). Acknowledgements We thank N. Whittle for comments on the manuscript.
20. Nelson, A. B. & Kreitzer, A. C. Reassessing models of basal ganglia function and C. Ramakrishnan and K. Deisseroth provided intersectional viral vectors. M. S.
dysfunction. Annu. Rev. Neurosci. 37, 117–135 (2014). Esposito and S. Arber provided flex-synaptophysin-GFP viral vectors. Z. Josh
21. Isosaka, T. et al. Htr2a-expressing cells in the central amygdala control Huang provided the SOM-Flp mouse line. We thank J. Eglinger for providing
the hierarchy between innate and learned fear. Cell 163, 1153–1164 overlap analysis scripts and S. Bourke for imaging advice. J.P.F. was funded
(2015). by an EMBO LTF (952-2011) and a NARSAD Young Investigator Fellowship.
22. Penzo, M. A. et al. The paraventricular thalamus controls a central amygdala C.X. was funded by EMBO ALTF (1579-2010). S.K., L.M. and P.T. were funded
fear circuit. Nature 519, 455–459 (2015). by a NARSAD Young Investigator Fellowship. All authors were supported by
23. Tovote, P. et al. Midbrain circuits for defensive behaviour. Nature 534, 206–212 the National Center of Competences in Research: ‘SYNAPSY — The Synaptic
(2016). Bases of Mental Diseases’ (financed by the Swiss National Science Foundation),
24. Beyeler, A. et al. Divergent routing of positive and negative information a SNSF core grant (to A.L.), an ERC Advanced Grant (to A.L.), and the Novartis
from the amygdala during memory retrieval. Neuron 90, 348–361 Research Foundation.
(2016).
25. Xu, C. et al. Distinct hippocampal pathways mediate dissociable roles of Author Contributions J.P.F. conceived, designed, performed and analysed
context in memory retrieval. Cell 167, 961–972.e16 (2016). most of the experiments and wrote the manuscript. S.K. conceived, designed,
26. Ciocchi, S., Passecker, J., Malagon-Vina, H., Mikus, N. & Klausberger, T. Brain performed and analysed all in vitro experiments. M.M. contributed to project
computation. Selective information routing by ventral hippocampal CA1 conceptualization, and performed and analysed rabies tracings. J.C. analysed
projection neurons. Science 348, 560–563 (2015). single-unit data in relation to behaviour. C.X. generated rabies tracing tools,
27. Bittencourt, A. S., Carobrez, A. P., Zamprogno, L. P., Tufik, S. & Schenberg, L. C. and performed and analysed rabies tracing experiments. L.M. performed the
Organization of single components of defensive behaviors within distinct overlap analysis. P.B. performed experiments. K.B., C.M. and A.K. performed
columns of periaqueductal gray matter of the rat: role of N-methyl-d-aspartic experiments and analysed histology. P.T. and A.L. conceptualized the project
acid glutamate receptors. Neuroscience 125, 71–89 (2004). and wrote the manuscript. All authors contributed to the experimental design
28. De Oca, B. M., DeCola, J. P., Maren, S. & Fanselow, M. S. Distinct regions of the and commented on the manuscript.
periaqueductal gray are involved in the acquisition and expression of defensive
responses. J. Neurosci. 18, 3426–3432 (1998). Author Information Reprints and permissions information is available at
29. Canteras, N. S. & Graeff, F. G. Executive and modulatory neural circuits of www.nature.com/reprints. The authors declare no competing financial
defensive reactions: implications for panic disorder. Neurosci. Biobehav. Rev. interests. Readers are welcome to comment on the online version of the
46, 352–364 (2014). paper. Correspondence and requests for materials should be addressed to
30. Pitman, R. K. et al. Biological studies of post-traumatic stress disorder. Nat. Rev. A.L. (andreas.luthi@fmi.ch) or P.T. (philip.tovote@fmi.ch).
Neurosci. 13, 769–787 (2012).
31. Paxinos, G. & Franklin, K. B. J. The Mouse Brain in Stereotaxic Coordinates. Reviewer Information Nature thanks K. Tye and the other anonymous
(Academic, 2001). reviewer(s) for their contribution to the peer review of this work.

0 0 M O N T H 2 0 1 7 | VO L 0 0 0 | NAT U R E | 5
© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

METHODS diameter, Thorlabs) two weeks after vector injection using the aforementioned
Animals. Male C57/Bl6J (Harlan/Envigo), Crf-ires-cre32, Som-ires-cre32, and methods. Optic fibre tips were lowered to 100–300 μ​m above the CEA. Implants
Pkcd-ires-cre11 mice aged 2–5 months were used for recording and behaviour. All were fixed to the skull with miniature screws (P.A. Precision Screws), cyanoacrylate
Cre driver lines were fully backcrossed to C57/Bl6J. For some in vitro experiments, glue (Ultra Gel, Henkel) and dental cement (Paladur, Heraeus). Mice had two
Crf-ires-cre mice were crossed to Som-ires-FlpO mice (Z. Josh Huang, Cold Spring additional weeks of recovery to allow for adequate opsin expression.
Harbour Laboratory) to generate CRF–Cre::SOM–Flp mice. For rabies tracing, For in vivo single-unit recordings, one hemisphere was implanted with an
Crf-ires-cre and Som-ires-cre mice were crossed to a LSL-R26Tva-lacZ33 line to optrode, consisting of an optic fibre with an attached 16-wire electrode, two
gene­rate CRF–Cre::LSL–R26Tva–lacZ (CRF–Cre–TVA) and SOM–Cre::LSL– weeks after vector injection. The electrodes tips were cut at an angle to protrude
R26Tva–lacZ (SOM–Cre–TVA) mice. All mice were individually housed on a 12 h approximately 300–500 μ​m beyond the fibre end. Electrodes were made of indi-
light/dark cycle during all experiments with ad libitum food and water. Behavioural vidually insulated, gold-plated nichrome wires (13 μ​m inner diameter, impedance
experiments were performed during the light cycle. All animal procedures were 70–100 kΩ​, Sandvik). The end of each wire was de-insulated and attached to an
performed in accordance with institutional guidelines and were approved by the 18-pin connector (Omnetics). The implant was fixed to the skull as described
Veterinary Department of the Canton of Basel-Stadt. above.
Conditioned flight paradigm. Two different contexts were used for all behavioural Viruses. For Cre-dependent expression of archaerhodopsin (Arch), we used
experiments. In context A, the mice were placed into a clear, cylindrical chamber rAAV2/5 CBA::flex–Arch–GFP. For Cre-dependent expression of channelrho-
with a smooth floor. The chamber was cleaned with 1% acetic acid. Context dopsin-2 (ChR2), we used rAAV2/5 EF1a::flex–ChR2(H134R)–eYFP. Control
B contained a clear, square enclosure with an electrical grid floor (Coulbourn mice for optogenetic experiments were mixed Cre+ mice from the Crf-ires-cre
Instruments) used to deliver footshock. This chamber was cleaned with 70% and Som-ires-cre colonies that were injected with rAAV2/1 CAG::flex–GFP. To label
ethanol. Both chambers contained an overhead speaker (model H12-01R, CRF+ somata for in vitro experiments, CRF–Cre::SOM–Flp mice were injected
Coulbourn Instruments) through which auditory stimuli were delivered at 75 dB. with rAAV2/1 CAG::flex–TdTomato. All of the above mentioned viruses were
Context B also contained a stimulus isolator (ISO-Flex, A.M.P.I.) for the delivery obtained from the University of Pennsylvania Vector Core. For Flp-dependent
of direct current (DC) shock. Auditory stimuli were generated using a System expression, we injected rAAVdj hSyn::CreOff/FlpOn–ChR2–eYFP34, or rAAV5
3 RP2.1 real time processor, an SA1 stereo power amp, and RPvdsEx software hSyn::CreOff/FlpOn–mCherry (Deisseroth laboratory, Stanford, produced by
(all Tucker-Davis Technologies). Behavioural protocols were generated with Vector Biolabs). For rabies tracing, custom-made rAAV2/7–EF1α​–flex–rabiesG
Radiant software (Plexon) to control all stimuli (CS, laser, shock and so on) via (UPenn Vector Core) and rabiesΔ​G–GFP–EnvA viruses (C. Xu, FMI) were used.
TTL pulses with high temporal precision. For axonal tracing experiments, rAAV2/9 CAG::flex–synaptophysin–GFP was
On day 1, following a 3 min baseline period, mice were presented with four used (S. Arber, FMI, produced by Vector Biolabs).
presentations of an auditory serial compound stimulus (SCS) of 20 s duration Optogenetic manipulations of behaviour. For optogenetic manipulations during
in context A, with a 60 s average pseudorandom ITI (range 50–90 s). The SCS behaviour, the implanted fibres were connected to a custom-built laser bench (Life
was comprised of a 10 s pure tone period (500 ms, 7.5 kHz pips at 1 Hz), followed Imaging Services) using an acousto-optic modulator (AA Opto-Electronic) to
immediately by a 10 s white noise period (500-ms pips at 1 Hz). The white noise control laser intensity (lasers: MBL473, 473 nm wavelength and MGL593.5,
was random and composed of frequencies from 1–20,000 Hz. On day 2 and day 3, 593.5 nm wavelength, CNI Lasers). The laser was connected from the port to the
after a 3 min baseline period, mice were conditioned in context B by five pairings implanted fibre via a custom-made patch cable (Thorlabs). Laser power at the
of the auditory stimulus with a 1 s, 0.9 mA DC footshock, with a 180 s average fibre tip was measured before every subject with an optical power and energy
pseudorandom ITI (range 150–210 s). The shock was applied immediately at the meter (PM100D, ThorLabs). Connectors were tested for coupling efficiency before
offset of the last pip. For retrieval tests, mice were placed back into context A and, implantations and laser power at the fibre tip for behavioural manipulations was
following a 3 min baseline period, were presented with the auditory stimulus four adjusted to reach an approximate irradiance value of 11 mW mm−2.
times with an average ITI of 90 s (range 60–120 s). Extinction of conditioned flight Cre+ mice of the same strain but from different litters were used in individual
behaviour was performed in context B. Following a 3 min baseline period, mice experiments. Mice were allocated to experimental groups without pre-determined
were presented with 16 presentations of the SCS with an average pseudorandom criteria and experimental subjects had unique identifiers that were later used to
ITI of 90 s (range 60–100 s). For the no shock control group, this paradigm was indicate genotype and group assignment. Optogenetic manipulations of condi-
unaltered except for the removal of the electric footshock on day 2 and day 3. In tioned behaviour were always performed with an internally controlled design in
initial experiments that led to the development of the SCS, the auditory stimuli which mice underwent eight trials, with alternating light-off and light-on con-
were either a pure 7.5 kHz tone or white noise. ditions. To inhibit CEA neuronal subpopulations during conditioned flight, the
Quantification of behaviour. All behavioural sessions were recorded to video yellow light was switched on 500 ms before the onset of the first white noise pip
using Cineplex software (Plexon) and mice were tracked using contour tracking. and remained on until the end of the last white noise pip (10 s in total). To inhibit
Freezing behaviour was scored from the video recording by an observer blind to SOM+ cells during the tone on day 3, the yellow light was switched on 500 ms
the condition, using the assistance of a frame-by-frame analysis of pixel changes before the onset of the first tone pip and was switched off immediately following
(Cineplex Editor, Plexon). By calibrating the known size of the chambers with the last tone pip (10 s total duration). For behavioural measurements in naive mice
the pixel dimensions of the camera to determine a calibration coefficient, speed of the Arch groups, the yellow light was switched on for 10 s, four times, with an
(cm s−1) was extracted using the animal’s centre of gravity. Jump escape behaviours ISI of 60 s. For ChR2 excitation of CRF+ cells, we used 10 ms pulses of blue light
were scored manually from video files. The flight score was calculated by dividing at 10 Hz. On day 3, the light was switched on 500 ms before the first pure tone pip
the average speed during each CS by the 10 s of pre-CS speed (speedCS/speedBL) and the pulse train continued for 10 s. On day 4, the light was switched on at the
and then adding 1 point for each escape jump. A flight score of 1 therefore indicates onset of the first pure tone pip and remained on for the entire SCS (20.5 s total).
no change from the pre-CS period. For ChR2 excitation of SOM+ cells, we used 5-ms pulses at 50 Hz (ref. 14). To test
Surgery. Mice were deeply anaesthetized with 5% isoflurane (Attane, Provet) in for effects in naive mice, the blue light pulse train (appropriate for each group) was
oxygen-enriched air (Oxymat 3, Weinmann), given an intraperitoneal injection on for 10 s, four times, with an ISI of 60 s.
of 1 mg kg−1 meloxicam (Metacam, Boehringer Ingelheim), and then fixed into Single-unit recordings. Single-unit spike sorting was performed using the Off-
a stereotaxic alignment instrument (Model 1900, Kopf Instruments). During Line Spike Sorter software (Plexon) as previously described16,35,36. In brief, princi-
surgery, mice were maintained on 1–2.5% isoflurane. Prior to scalp incision, a local pal components were calculated for unsorted waveforms and plotted in principal
injection of 0.1 ml ropivacain (Naropin, AstraZeneca) was made subcutaneously. component space. Clusters were manually defined, template waveforms were saved,
Mice were maintained at a core temperature of 36 °C using a feedback-controlled and template-matching was used to verify unit identity across sessions. Sort quality
DC temperature controller (FHC). was quantified using the J3 statistic and Davies–Bouldin validity index, as previ-
For selective expression of inhibitory or excitatory opsins, Cre-dependent ously described16,35,36. Additionally, we determined unit stability by quantifying the
recombinant adeno-associated viral vectors (AAV) were injected into the central similarity of waveform shapes using linear correlation (r) values16,35,36.
amygdala (CEA). AAVs (approximately 0.25 μ​l per hemisphere) were delivered Neuronal activity is reported either as frequency or normalized activity (z-score
into the CEA using pulled glass pipettes (tip diameter 10–20 μ​m) connected to a transformation). z scores used to determine CS-induced neuronal activity changes
Picospritzer III microinjection system (Parker Hannifin Corporation). CEA coor- were calculated for each neuron by subtracting the population mean (the average
dinates were: 1.2–1.3 mm posterior to bregma, ±​2.9–3.0 mm lateral to the midline baseline firing rate in the 300 ms pre-stimulus period across all trials and pips) from
and 3.9–4.1 mm below the cortical surface. each individual raw value (50-ms bins across all trials and pips), and then dividing
For optogenetic manipulations of behaviour, animals were bilaterally implanted this difference by the standard deviation of the population mean. z scores for each
with custom-built optic fibre connectors (fibre: 0.48 numerical aperture, 200 μ​m neuron were then averaged across cells for each population. Any statements of

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

significance were made only if the average activity of an identified population Immunohistochemistry related to in vitro physiology. After fixation for 1 h in
reached a ±​1.645 change in z score for at least one bin during the period of interest. 4% paraformaldehyde in PBS, 300-μ​m brain slices were stored in PBS overnight
To evaluate unit activity relative to behaviour, we normalized to the episodes of (4 °C). The next day, free-floating sections were washed with PBS three times before
freezing and flight behaviours. Because the duration of those behaviours is variable, incubation in blocking solution (3% normal goat serum (vol/vol), 1% bovine serum
the time periods before, during, and after freezing and flight behaviours were each albumin (BSA, vol/vol) and 0.5% Triton X-100 (vol/vol) in PBS) for 2–3 h at room
divided into equal numbers of bins and spikes were sorted into these bins. The temperature (20–22 °C). Afterwards, slices were incubated for 48 h at 4 °C in carrier
resulting activity was then normalized to the pre-freezing or pre-flight period solution (same as above) with a combination of the following antibodies: rat anti-
using a z-score transformation. SOM (1:500; Millipore, MAB354), mouse anti-PKCδ​(1:500; BD Biosciences,
Optical identification of units. For optogenetic identification of CRF+ or SOM+ 610398), rabbit anti-GFP (1:1,000; Life Technologies, A11122), chicken anti-RFP
neurons, we used pulses of either blue or yellow light (to activate ChR2 or Arch, (1:500; Millipore, AB3528), mouse anti-bassoon (1:500, Enzo Life Sciences, ADI-
respectively). We used 300-ms pulses, 60–120 times, with a 2 s inter-pulse interval, VAM-PS003). Sections were washed with 0.1% Triton X-100 in PBS three times
at 10–20 mW light power at the fibre tip. Units were considered as light responsive before adding secondary antibodies (goat anti-rat Alexa Fluor 647, 1:500, A21247;
if they showed significant, time-locked (<​10 ms) changes in neuronal activity upon goat anti-mouse Alexa Fluor 647 or 568, 1:750, A31625 or A11031; goat anti-rabbit
illumination36. For units identified with ChR2, we calculated the jitter of first light- Alexa Fluor 488, 1:750, A27034; goat anti-chicken Alexa Fluor 568, 1:500, A11041;
evoked spikes by calculating the difference between samples and then dividing by all Life Technologies) and streptavidin conjugated to Alexa Fluor 405 (1:1,000;
the number of samples (minus 1). To determine the onset of inhibition in neu- Life Technologies, S32351) in carrier solution for 24 h at 4 °C. Finally, slices were
rons with low baseline firing rates, we used change-point analysis (Change Point washed in PBS four times, mounted and coverslipped, and imaged using a confocal
Analyzer 2.0, Taylor Enterprises Inc.). As described previously36, this identifies microscope (LSM710, Carl Zeiss AG) with 63×​objective and twofold digital zoom.
the time point exhibiting a significant change in neuronal activity relative to the Only reconstructed cells were included in the data analysis. Recordings from cells
preceding time points. For all identified units, we calculated linear correlation (r) immuno-negative for SOM/mCherry, PKCδ​or tdTomato were summarized in an
values for spontaneous and light-evoked spikes to quantitatively determine the ‘unidentified’ group.
similarity of their waveform shapes. Note that for two SOM+ cells identified using Histology and immunohistochemistry for optogenetics and single-unit recordings.
Arch, there were not enough spikes inside the light period to calculate the r value. Mice were deeply anaesthetized with tribromoethanol (Avertin; 0.3 g kg−1) and an
In vitro electrophysiology. Four weeks after virus delivery, coronal brain slices electrolytic lesion was made at the electrode tip by applying 0.1 μ​A for 10 s (with a
were prepared from CRF-ires-cre or CRF– Cre::SOM –Flp mice injected with rAAV polarity switch) to one of the electrode wires. Mice were then perfused transcardially
2/5 EF1a::flex–ChR2(H134R) –eYFP or rAAVdj hSyn::CreOff/FlpOn–ChR2–eYFP with PBS followed by 4% paraformaldehyde in PBS. Brains were removed and
and rAAV2/1 CAG::flex–tdTomato, respectively. For some experiments, CRF– post-fixed overnight at 4 °C. 80-μ​m coronal brain slices were cut with a vibratome
Cre::SOM–Flp mice injected with rAAV 2/5 EF1a::flex–ChR2(H134R) –eYFP (VT1000 S, Leica) and stored in PBS. Standard immunohistochemical procedures
and rAAV5 hSyn::CreOff/FlpOn–mCherry were used. Mice were deeply anaes- were performed on free-floating brain sections. Sections were blocked in 5% normal
thetized by intraperitoneal injection of ketamine/medetomidine (250 mg kg−1 goat serum (in 0.3% Triton X-100 in PBS; NGS) for 2 h at room temperature
and 2.5 mg kg−1 bodyweight, respectively) and transcardially perfused with ice- (20–22 °C), followed by overnight incubation with primary antibodies (rabbit anti-
cold ACSF (in mM: 124 NaCl, 2.7 KCl, 26 NaHCO3, 1.25 NaH2PO4, 2.5 glucose, GFP, 1:1,000; Life Technologies, A11122) in NGS at 4 °C. The next day, slices were
50 sucrose, 0.1 CaCl2, 6 MgCl2, 3 kynurenic acid, oxygenated with 95% O2/5% washed 3×​for 10 min in 0.3% Triton X-100, and then incubated with secondary
CO2, pH 7.4). The brain was quickly removed from the skull and coronal brain antibodies (goat anti-rabbit Alexa Fluor 488, 1:1,000, A27034; Life Technologies)
slices (300 μ​m) containing the CEA were cut in ice-cold perfusion ACSF using in NGS for 2 h at room temperature. Slices were washed for 10 min in PBS, given a
a vibratome (HM 650 V, Microm) equipped with a sapphire blade. Brain slices 10 min exposure to 4′​,6-diamidin-2-phenylindol (DAPI, 1:10,000, Sigma-Aldrich),
were allowed to recover in an interface chamber with recording ACSF (in mM: and then washed three times for 15 min in PBS. Slices were mounted on gelatin-
126 NaCl, 2.7 KCl, 26 NaHCO3, 1.25 NaH2PO4, 18.6 glucose, 2.25 ascorbic acid, coated glass slides, coverslipped and imaged using an Axioscan Z1 (Carl Zeiss
2 CaCl2, 1.3 MgCl2, oxygenated with 95% O2/5% CO2, pH 7.4) for 45 min at 37 °C AG). To quantify the number of opsin-expressing somata in the CEl versus CEm,
and subsequently kept at room temperature (20–22 °C) until start of recordings. three sections around the optic fibre tip in each hemisphere were scanned with
Sections were transferred to a submerged recording chamber on an upright a 10×​air objective using a laser-scanning confocal microscope (LSM700, Carl
microscope (BX50WI, Olympus) and constantly superfused with recording ACSF Zeiss AG). z stacks of multi-tiled regions of interest were acquired. Manual cell
(2–4 ml min−1; as above, except: 2.5 mM CaCl2, 10 μ​M CNQX (6-cyano-7-nitro- counting of fluorophore-expressing somata was then performed and values were
quinoxaline-2,3-dione), 10 μ​M CPP (3-((R)-2-carboxypiperazin-4-yl)-propyl-1- averaged.
phosphonic acid)) at 35 ±​ 1 °C. eYFP- and tdTomato/mCherry-expressing neurons Fibre tips and electrode lesions were located and matched against a mouse brain
were visualized using epifluorescence and a 40×​objective (LumPlanFl 40×​/0.8, atlas (Paxinos and Franklin, 2001). Mice were included into the behavioural analysis
Olympus). if they had bilateral expression in the CEA and if the fibre tips were no more than
Voltage clamp recordings from CEA neurons were acquired in whole-cell mode 300 μ​m away from the CEA.
at a holding potential of −​70 mV using 3–5 MΩ​glass pipettes filled with inter- Overlap analysis. To detect overlap between CRF+ and PKCδ​+ or SOM+ neurons,
nal solution (in mM: 110 CsCl, 30 K-gluconate, 1.1 EGTA, 10 HEPES, 0.1 CaCl2, CRF-ires-cre mice were injected with rAAV2/1 CAG::flex–GFP to mark CRF+
4 Mg-ATP, 0.3 Na-GTP, 4 QX-314 chloride and 0.4% biocytin, pH 7.3). ChR2- somata. For quantification, serial vibratome sections (80 μ​m) were obtained from
expressing neurons were photostimulated using a blue LED (465 nm, Plexon, LED- two and three animals for PKCδ​and SOM, respectively. PKCδ​+ or SOM+ cells
driver LD-1) connected to an optical fibre, which was positioned above the CEA. were visualized by immunostaining using a primary antibody against PKCδ​ raised
Blue light pulses of 10 mW with 10 ms duration were applied at a frequency of 1 Hz. in mouse (1:500, BD Transduction Laboratories, 610398) and SOM raised in rat
In some slices, 100 μ​M picrotoxin was administered with the recording ACSF for (1:500, Millipore, MAB354). They were detected by Alexa Fluor 647-conjugated
the last cell recorded from. Spike suppression was tested in current clamp mode secondary antibodies (1:1,000, Invitrogen, goat anti-mouse Alexa Fluor 647,
(internal solution in mM: 130 K-methylsulfate, 10 HEPES, 10 Na-phosphocreatine, catalogue number A-20990 and goat anti-rat Alexa Fluor 647, catalogue number
4 Mg-ATP, 0.3 Na-GTP, 5 KCl, 0.6 EGTA and 0.4% biocytin, pH 7.3). Spikes were A-21248). For image acquisition, sections (PKCδ​, n =​  4; SOM, n =​ 6) were scanned
evoked from a holding potential of −​60 mV with current injections of 20–60 pA with a 20×​air objective (Plan-Apochromat 20×​, NA  =​ 0.8) using a laser-scan-
for 500 ms, and the same current step was subsequently paired with blue light ning confocal microscope (LSM700, Carl Zeiss AG). z stacks of multi-tiled regions
stimulation to activate ChR2-expressing neurons. Spike probability of individual of interest were then acquired. Individual tiles were stitched using the stitching
neurons with and without blue light stimulation was calculated from ten repeated plugin37 in Fiji (ImageJ software) and downsampled (factor: 0.5). After down-
trials, except for one SOM+ cell in which there were nine repeated trials. Data were sampling, we applied a segmentation classifier using the 3D Weka segmentation
acquired with a Multiclamp 700A amplifier, Digidata 1440A A/D converter and classifier trained in Fiji and probability maps were obtained. The probability maps
pClamp 10 software (all Axon instruments) at 20 kHz and filtered at 4 kHz (voltage were then converted into cell segmentation masks using a Python script in Fiji.
clamp) or 10 kHz (current clamp). Series resistance was monitored continuously The intensity of each individual cell was measured using the 3D manager plugin38.
and experiments were discarded when exceeding 30 MΩ​. In each slice, 2–6 CEA Monosynaptic rabies tracing of inputs to CRF + and SOM + neurons.
neurons surrounded by ChR2–eYFP-positive fibres were recorded and filled with A mixture of rabies∆​G –mCherry–EnvA, or rabies∆​ G –GFP–EnvA, and
biocytin for post hoc immunohistochemical identification of cell type after elec- AAV2/7–flex–rabiesG (3:2; 500 nl total volume) was injected into the CEA
trophysiology. Outside-out patches were pulled at the end of each recording and of CRF–Cre–TVA and SOM–Cre–TVA mice. In other animals, we first
slices were fixed for 1 h in 4% paraformaldehyde in PBS for subsequent immu- injected AAV2/7–flex–rabiesG, followed by an injection of rabies∆​G–GFP–
nohistochemistry. All chemicals were purchased from Sigma-Aldrich except for EnvA a week later. As the two strategies yielded comparable results, the data
CNQX, CPP and QX-314 chloride (from Tocris Bioscience). were pooled together. One week after the rabies virus was injected, mice

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

were transcardially perfused with PBS, followed by 4% paraformaldehyde DAPI counterstain, as described above. Images were acquired with a laser-scanning
in PBS. Brains were post-fixed overnight at 4 °C. Brains were cut into 80-μ​m confocal microscope (LSM700, Carl Zeiss AG).
coronal slices with a vibratome (Leica). Free-floating sections were rinsed in PBS. Statistics. All data were statistically analysed using Prism 6 (GraphPad Software,
Sections were then incubated in blocking solution (3% bovine serum albumin and Inc.). All data were checked for normal distribution using the Shapiro–Wilk
0.5% Triton X-100 in PBS; BSA) containing a guinea pig anti-rabies glycoprotein normality test (α =​ 0.05), and the appropriate parametric, or non-parametric,
(rabG) antibody for 48 h at 4 °C (1:500, custom order, Eurogentec). Subsequently, test for significance was run. No statistical methods were used to predetermine
sections were washed with PBS three times for 5 min and then incubated for 4 h at sample size.
room temperature with goat anti-guinea pig Alexa Fluor 647 (1:500; Invitrogen, Data availability. The data sets generated during and/or analysed during the
A-21450) in BSA. Finally, immunolabelled sections were rinsed three times with current study are provided as Source Data, or are available from the corresponding
PBS, mounted on gelatin-coated slides, dehydrated and coverslipped. Individual author on reasonable request.
brain slices were imaged with a LSM700 confocal microscope (Carl Zeiss AG).
Quantitative analysis of rabies tracing results. Brain sections were scanned by an
32. Taniguchi, H. et al. A resource of Cre driver lines for genetic targeting of
AxioScan (Carl Zeiss AG) with a 5×​objective. Images for individual brain sections GABAergic neurons in cerebral cortex. Neuron 71, 995–1013 (2011).
were sorted in the correct order, aligned with the TrackEM plugin for Fiji (Fiji 1.48), 33. Seidler, B. et al. A Cre-loxP-based mouse model for conditional somatic gene
and then exported into Imaris software (Imaris 7.3.1) for analysis. The contours expression and knockdown in vivo by using avian retroviral vectors. Proc. Natl
for different brain structures were manually drawn in Imaris using a mouse brain Acad. Sci. USA 105, 10137–10142 (2008).
atlas for reference. Somata were semi-automatically detected as spots in Imaris 34. Fenno, L. E. et al. Targeting cells with single vectors using multiple-feature
Boolean logic. Nat. Methods 11, 763–772 (2014).
(automated detection followed by manual verification to eliminate false positive 35. Herry, C. et al. Switching on and off fear by distinct neuronal circuits. Nature
and false negative results). Finally, the total number of cells for a given brain area 454, 600–606 (2008).
was exported from Imaris and quantified. 36. Wolff, S. B. et al. Amygdala interneuron subtypes control fear learning through
Anterograde tracing of axons from CRF+ neurons. To determine projection disinhibition. Nature 509, 453–458 (2014).
targets of CRF+ neurons, CRF-ires-cre mice were injected into the CEA with an 37. Preibisch, S., Saalfeld, S. & Tomancak, P. Globally optimal stitching of
tiled 3D microscopic image acquisitions. Bioinformatics 25, 1463–1465
AAV carrying a Cre-conditional synaptophysin conjugated to GFP to observe
(2009).
axon terminals (S. Arber, FMI). Mice were perfused, and tissue was collected and 38. Ollion, J., Cochennec, J., Loll, F., Escudé, C. & Boudier, T. TANGO: a generic tool
processed as described above (see Histology and immunohistochemistry for opto- for high-throughput 3D image analysis for studying nuclear organization.
genetics and single-unit recordings). Sections were stained against GFP, with a Bioinformatics 29, 1840–1841 (2013).

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 1 | See next page for caption.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

Extended Data Figure 1 | Data from all mice presented in Fig. 1, and flight in response to a single CS. e, Two groups of mice were subjected to
single CS conditioned flight and no-shock control groups. a, The data the flight paradigm and either a pure tone (n =​ 10) or white noise (n =​  10)
from Fig. 1d (day 3, conditioning) represented as a line plot to illustrate was paired with a 1 s shock during conditioning. f, Flight scores for each
changes in each mouse. Left, the number of escape jumps (n =​  20, group, across days. While active behaviour developed in both groups, the
Friedman test, P <​ 0.0001, Dunn’s multiple comparisons test). Middle, flight response was significantly greater in the white noise group on day
flight scores for each mouse. The pre-SCS period is used to calculate 3 on the first two trials (two-way repeated-measures ANOVA, cue ×​ trial
the flight score and is therefore plotted as a value of 1 (n =​  20, Wilcoxon interaction, F(17,153) =​  1.81, P <​ 0.05, Bonferroni’s multiple comparison
matched-pairs signed-rank test). Right, freezing values (n =​  20; one-way test). g, Freezing responses for each group, across days. Freezing in
repeated-measures ANOVA, F =​  56.82, P <​ 0.0001, Tukey’s multiple the pure tone or white noise group was similar on all trials (two-way
comparisons test). b, The data from Fig. 1e (day 4, extinction) represented repeated-measures ANOVA, cue ×​trial interaction, F(17,153) =​  1.62,
as a line plot to illustrate changes in each mouse. Left, flight scores for P =​  0.06). h, Schematic of behavioural paradigm used to test for possible
each mouse. The pre-SCS period is used to calculate the flight score aversive nature of white noise. i, The SCS was presented but was never
and is therefore plotted as a value of 1 (n =​ 12; Wilcoxon matched-pairs paired with footshock. j, Left, flight was not observed in the conditioning
signed-rank test). Right, freezing values, including the pre-SCS period, context in the absence of pairing with footshock (n =​  10, two-tailed
during the first block of four trials of extinction (n =​ 12; Friedman test, paired t-test). Right, freezing values remained at low baseline values in
P <​ 0.01, Dunn’s multiple comparisons test). c, The data from Fig. 1f (day the absence of pairing with footshock (n =​ 10, Friedman test). k, Left,
4, retrieval) represented as a line plot to illustrate changes in each mouse. flight was not observed in the neutral context (n =​ 10, two-tailed paired
Left, flight scores for each mouse. The pre-SCS period is used to calculate t-test). Right, freezing values were decreased, not increased, in response
the flight score and is therefore plotted as a value of 1 (n =​  8; two-tailed to the SCS (n =​ 10, one-way ANOVA, F =​  9.45, P <​ 0.01, Tukey’s multiple
paired t-test). Right, freezing values, including the pre-SCS period (n =​  8; comparisons test). Horizontal lines denote the mean. Values in f and g are
one-way repeated-measures ANOVA, F =​  49.55, P <​  0.0001, Tukey’s means ±​  s.e.m. *​P <​  0.05; *​*​P <​  0.01.
multiple comparisons test). d, Schematic of the paradigm used to elicit

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 2 | See next page for caption.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

Extended Data Figure 2 | Data related to Fig. 2, and loss-of-function for protocol). f, Average speed traces from naive mice given four, 10-s light
PKCδ groups, effects of optical inhibition on naive mice, and overlap stimulations (GFP control, n =​  8;CRF–Arch, n =​  7; SOM–Arch, n =​  11;
analysis. a, Positions of fibre tips for loss-of-function experiments. PKCδ​–Arch, n =​  11). g, Flight and freezing values for all mice shown in
Placements were determined using standard histological techniques Fig. 2, plus the PKCδ​–Arch group (flight scores: two-tailed paired t-test
and lesions were matched to a mouse brain atlas. Mouse brain figure for GFP control, SOM–Arch, and PKCδ​–Arch; Wilcoxon matched-pairs
reproduced with permission from ref. 31. b, Injection and implantation signed-rank test for CRF–Arch; freezing scores: Wilcoxon matched-pairs
strategy for identified single-unit recordings used to demonstrate Arch- signed-rank test for GFP control and SOM–Arch; two-tailed paired t-test
mediated inhibition. c, Single-unit recordings from identified CEl units for CRF–Arch and PKCδ​–Arch). Horizontal lines indicate the mean.
in behaving mice. Units were isolated and identified as described (Fig. 3, h, Flight and freezing values for naive mice given four, 10-s light
Extended Data Fig. 3 and Methods). The light was switched on four times stimulations (Wilcoxon matched-pairs signed-rank test for freezing
for 10 s. Shown for each group is an example raster above and the average values). Horizontal lines indicate the mean. i, Example images from
peri-event histogram of firing rate below. The light inhibited neuronal overlap analysis between CRF+ neurons and SOM+ or PKCδ​+ neurons in
activity in all three cell types. d, Expression of Arch in CEl PKCδ​+ neurons. the CEA. Arrowhead indicates a rare example of overlap between CRF+
Bregma −​1.34 mm caudal. Scale bar, 250 μ​m. e, Average speed traces of and PKCδ​+. Scale bar, 10 μ​m. j, Percentage of overlap. (PKCδ​+, n =​  4
PKCδ​–Arch mice (n =​ 8) in the conditioned flight paradigm, illustrating sections from two mice; SOM+, n =​ 6 sections from three mice). Values are
that there was no effect of the light on conditioned flight (see Fig. 2 for means ±​  s.e.m. *​P <​  0.05.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 3 | See next page for caption.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

Extended Data Figure 3 | Data related to Fig. 3, and neuronal responses after light pulses. e–j, Normalized activity (z score, bottom) and example
of identified units to tone on day 3. a, Positions of electrode lesions raster plots (top) of CEl CRF+ or SOM+ neurons to the tone component
and fibre tips for data presented in Fig. 3. Electrolytic lesions were made of the SCS. e, Neuronal response to the tone component for CRF+ neurons
at the tip of one wire (see Methods) and placements were determined (n =​ 8) during pre-exposure (day 1). Pre-SCS firing rate: 4.5 ±​  1.3 spikes
using standard histological techniques. Lesions were matched to a per s. f, CRF+ neurons were not significantly activated by the pure tone on
mouse brain atlas. Mouse brain figure reproduced with permission day 3 of the flight paradigm (n =​ 8). Pre-SCS firing rate: 5.5 ±​ 2 spikes per s.
from ref. 31. b, Raster plots and peri-event histograms of firing rate g, There was no significant activation of CRF+ cells to the tone in the
for example neurons from each condition illustrating Arch-mediated neutral context (n =​ 7). Pre-SCS firing rate: 5.4 ±​ 2.3 spikes per s. h, SOM+
inhibition, or ChR2-mediated excitation, of CEl CRF+ and SOM+ cells neuronal activity (n =​ 10) was not significantly changed in response to the
(an example from the CRF–ChR2 group is presented in Fig. 3). c, Latency tone during the pre-exposure session (day 1). Pre-SCS firing rate: 2.7 ±​  0.6
to activation/inhibition plotted against z scores for all CRF+ and SOM+ spikes per s. i, Neuronal responses of SOM+ neurons to the tone on day 3
neurons. Average latency: CRF+ =​  5.25  ±​ 0.88 ms; SOM+ =​  4.6  ±​ 0.9 ms. of the flight paradigm (n =​ 10). Pre-SCS firing rate: 5.8 ±​ 1.3 spikes per s.
d, Left, waveforms of action potentials were unchanged by light for all These higher firing rates are consistent with the pre-SCS contextual
unidentified units recorded in Arch-expressing mice with identified units. freezing observed on this day. j, SOM+ cells (n =​ 8) are excited by the tone
Centre, firing rates of unidentified units before, during, and after light, in the neutral context. Pre-SCS firing rate: 2.1 ±​ 0.4 spikes per s. Values are
illustrating that the light pulses had no effect on unidentified unit activity. means ±​  s.e.m.
Right, z-score transformation of unidentified unit activity during and

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 4 | See next page for caption.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

Extended Data Figure 4 | Data related to Fig. 4, and effects of optical CRF–ChR2 (right) mice shown in Fig. 4d (two-tailed paired t-test for
excitation on naive mice and CRF–ChR2 mice during retrieval. CRF control freezing, CRF–ChR2 flight, CRF–ChR2 freezing. Wilcoxon
a, Top, positions of fibre tips for CRF+ and SOM+ gain-of-function matched-pairs signed-rank test for CRF control flight). Horizontal lines
groups. Placements were determined using standard histological indicate the mean. f, Flight and freezing values for individual SOM control
techniques and lesions were matched to a mouse brain atlas. Mouse brain (left) and SOM–ChR2 (right) mice shown in Fig. 4d (two-tailed paired
figure reproduced with permission from ref. 31. b, Single-unit recordings t-test). Horizontal lines indicate the mean. g, The CRF–ChR2 (n =​  7)
from identified CEl units in behaving mice. Units were isolated and and GFP control (n =​ 10) groups were subjected to the conditioned flight
identified as described (see Fig. 3, Extended Data Fig. 3 and Methods). paradigm. They were placed into the neutral context on day 4 (retrieval)
The light was switched on four times for 10 s (see Methods). Shown for and the SCS was played eight times. On four of the trials, the blue light
each group is a raster plot of an example neuron above and a peri-event was turned on during the entire SCS. Left, flight and freezing values
histogram of the average firing rate below. The light protocols were for individual mice in the CRF control group (flight scores: Wilcoxon
effective in exciting both neuronal subclasses. c, Average speed traces matched pairs signed-rank test; freezing scores: two-tailed paired t-test).
from naive mice given four, 10-s light stimulations (CRF control, n =​  10; Middle, flight and freezing values for individual mice in the CRF–ChR2
CRF–ChR2, n =​ 7; SOM control, n =​  7; SOM–ChR2, n =​ 7). Blue light group (two-tailed paired t-test). Horizontal lines indicate the mean.
caused a decrease in speed in the SOM–ChR2 group. d, Flight and freezing Right, average change in freezing and flight between light and no-light
values for naive mice given four, 10-s light stimulations. ChR2-mediated conditions. CRF+ excitation significantly reduced conditioned freezing
excitation of SOM+ cells induced freezing behaviour (two-tailed paired (two-tailed unpaired t-test). Box and whisker plots indicate median,
t-test, CRF–ChR2 and SOM–ChR2; Wilcoxon matched-pairs signed-rank interquartile range, and 5th to 95th percentiles of the distribution. Crosses
test, CRF control and SOM control). Horizontal lines indicate the mean. indicate means. All other values are means ±​ s.e.m., except as indicated.
e, Flight and freezing values for individual CRF control (left) and

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 5 | In vitro slice recording data related to Fig. 4, +


(n =​ 7) or unidentified (n =​ 8) cells (Kruskal–Wallis test). k, Average
and bassoon immunolabelling to determine monosynaptic connectivity latency to IPSC peak from start of photostimulation (Kruskal–Wallis test
between CRF+ and SOM+ cells. a, Top, a light-evoked action potential P <​ 0.05; Dunn’s multiple comparisons test, no significant differences).
recorded in cell-attached mode from an example ChR2-expressing CRF+ l, Average rise time of IPSCs (10–90%). No significant differences were
cell. The average latency to light-evoked spike was 3.3 ±​ 2.2 ms (n =​  4). observed (Kruskal–Wallis test). m, Whole-cell recordings from CEl
Bottom left, ChR2 expression in the example CRF+ cell. Bottom right, SOM+ cells were performed while stimulating ChR2-expressing CRF+
recorded neurons were filled with biocytin after recording. b, Whole- cells with blue light. n, Top, IPSCs evoked in a SOM+ neuron by brief
cell recordings from CEl target cells were performed while stimulating photostimulation of the CRF+ network (individual traces from one cell in
ChR2-expressing CRF+ cells with blue light. c, Top left, example histology grey, corresponding average IPSC in black). Bottom, IPSCs were blocked
showing ChR2–eYFP fibres from CRF+ cells and biocytin-filled cells from by application of the GABAA-receptor antagonist picrotoxin. o, Whole-cell
whole-cell patch-clamp recording. Top right, IPSCs evoked in a PKCδ​+ recordings from CEl CRF+ cells were performed while stimulating ChR2-
neuron by brief photostimulation of the CRF+ network (individual traces expressing SOM+ cells with blue light. p, Top, IPSCs evoked in a CRF+
from one cell in grey, corresponding average IPSC in black). Bottom, neuron by brief photostimulation of the SOM+ network (individual traces
immunohistochemistry performed after electrophysiology recordings from one cell in grey, corresponding average IPSC in black). Bottom,
revealed PKCδ​ expression. d, Average IPSC amplitudes normalized to IPSCs were blocked by application of the GABAA-receptor antagonist
cellular capacitance as approximation for cell size. e, Average latency picrotoxin. q, The amplitude of light-evoked IPSCs was significantly
to IPSC peak from start of photostimulation. f, Average rise time of reduced by application of picrotoxin for both
IPSCs (10–90%). For d–f, no significant differences were observed CRF to SOM (n =​ 3; reduction to 3.9% of baseline amplitude) and SOM
between SOM+ (n =​  23), PKCδ​+ (n =​ 8) or unidentified (n =​  13) cells to CRF (n =​ 4; 2.8% of baseline) connectivity (two-tailed paired t-test).
(Kruskal–Wallis test). g, Top, a light-evoked action potential recorded r, Left, mCherry label of a SOM+ cell which received inhibitory input from
from a ChR2-expressing SOM+ cell. The average latency to light- the CRF+ network. Middle, CRF–ChR2+ fibres come in close apposition
evoked spike was 3.0 ±​ 1.1 ms (n =​ 3). Bottom left, ChR2 expression (arrowheads) to the biocytin-filled SOM+ cell (maximum intensity
in the example SOM+ cell. Bottom right, recorded neurons were filled projection). Right, single confocal planes of appositions labelled with
with biocytin after recording. h, Whole-cell recordings from CEl target arrowheads in middle panel. Co-immunolabelling with the presynaptic
cells were performed while stimulating ChR2-expressing SOM+ cells marker bassoon suggests monosynaptic connections between CRF+ and
with blue light. i, Top left, example histology showing ChR2–eYFP SOM+ neurons. s, Left, tdTomato labelling of a CRF+ cell which received
fibres from SOM+ cells and biocytin-filled cells from whole-cell patch- inhibitory inputs from the SOM+ network. Middle, SOM–ChR2+ fibres
clamp recordings. Top right, IPSCs evoked in a PKCδ​+ cell by brief come in close apposition (arrowheads) to the CRF+ cell. Right, single
photostimulation of the SOM+ network. Bottom, immunohistochemistry confocal planes of appositions labelled with arrowheads in middle panel.
performed after electrophysiology recordings revealed PKCδ​ expression. Co-immunolabelling with the presynaptic marker bassoon suggests
j, Average IPSC amplitudes normalized to cellular capacitance. No monosynaptic connections between SOM+ and CRF+ neurons. Horizontal
significant differences were observed between CRF+ (n =​  24), PKCδ​ lines throughout denote the mean. *​*​*​P <​  0.001.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH LETTER

a b
Rabies
AAV-flex-RabiesG and CEl CEm
RabiesΔG-mCherry-EnvA
CEl

BLA
CEm

BLA Glycoprotein
CEA
CRF-Cre-TVA or BLA CEl
CEm
SOM-Cre-TVA mice

BLA
d CRF SOM
IC
PVT c
BLA
vHP
vHIPP
BLP BLP
Apir PVT

40 20 0 20 40
% Input cells/total presynaptic cells APir

e
Target Target Number Number of Number of presynaptic cells
site cell type of mice starter cells Apir vHP BLA BLP PVT IC

375 812.3 628.7 645.3 560.7 335.3 663.7


CEA SOM+ 3
±49.7 ±237.7 ±181.2 ±151.4 ±172.7 ±139.7 ±201.1

15.8 140.0 55.5 28.8 114.8 37.5 22.0


CEA CRF+ 4
±4.3 ±62.6 ±31.1 ±7.0 ±60.8 ±23.6 ±10.0

f g h
AAV-flex-synaptophysin-GFP
CEl PH

LH
PeF
CEm
DM

MTu VMH
BLA
CEA BLA

Arc
100µm
µm 200µm
CRF-IRES-Cre
mouse

i j k
dmPAG

dl/lPAG
dl/lPAG
VMH

vlPAG
vlPAG
Arc

100µm 200µm 100µm

Extended Data Figure 6 | See next page for caption.

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
LETTER RESEARCH

Extended Data Figure 6 | Input structures of CRF+ and SOM+ neurons, projecting to CRF+ cells (repeated-measures one-way ANOVA F =​  14.39,
and axonal targets of CRF+ cells. a, Pseudotyped-rabies tracing approach P <​ 0.05). APir, amygdalopiriform transition area; BLA, basolateral
used to identify input areas to the CEA CRF+ or SOM+ population. amygdala; IC, insular cortex. e, Table listing all raw values from rabies-
A Cre-conditional AAV encoding the rabies glycoprotein and a G-deleted tracing experiments. f, Injection strategy used to locate terminal fields
EnVA-pseudotyped rabies were injected into the CEA of mice. Mouse of CEA CRF+ axonal projections. A Cre-conditional AAV encoding
brain figure reproduced with permission from ref. 31. b, Left, merged synaptophysin conjugated to GFP was injected into the CEA of CRF-ires-
image of glycoprotein and rabies expression localized to CRF+ cells of cre mice. Mouse brain figure reproduced with permission from ref. 31.
the CEA. Arrowheads indicate starter cells, which co-express rabies g, Expression of GFP in the CEA illustrating transfected somata and GFP+
and glycoprotein. Inset, an example starter cell expressing both rabies puncta representing local axon terminals. h, CRF+ neurons project to
and glycoprotein. Top right, rabies expression in the CEA. Bottom multiple structures in the hypothalamus. PH, posterior hypothalamic
right, neurons expressing glycoprotein. c, Example regions providing area; LH, lateral hypothalamic area; PeF, perifornical nucleus; DM,
monosynaptic input to the CRF+ network. vHIPP, ventral hippocampus; dorsomedial hypothalamic nucleus; MTu, medial tuberal nucleus; VMH,
BLP, basolateral amygdaloid nucleus, posterior part; PVT, paraventricular ventromedial hypothalamic nucleus; Arc, arcuate hypothalamic nucleus.
thalamic nucleus. d, Comparison of the number of presynaptic cells to i, Demonstration of CRF+ terminals within the VMH. j, CEA CRF+
CRF+ and SOM+ cells, normalized to the total number of presynaptic cells neurons also project to the peri-aqueductal grey (PAG). dmPAG,
in the defined regions. While there are a similar number of presynaptic dorsomedial PAG; dl/lPAG, dorsolateral/lateral PAG; vlPAG, ventrolateral
neurons across brain regions projecting to SOM+ cells (Friedman test, PAG. k, CEA CRF+ cells project widely throughout PAG columns.
P =​ 0.32), there are significantly different numbers of presynaptic neurons

© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

Das könnte Ihnen auch gefallen