Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Articles

https://doi.org/10.1038/s41593-020-00728-x

Tau accumulation in astrocytes of the dentate


gyrus induces neuronal dysfunction and memory
deficits in Alzheimer’s disease
Kevin Richetin 1,2,3 ✉, Pascal Steullet1, Mathieu Pachoud2,3, Romain Perbet 4, Enea Parietti1,
Mathischan Maheswaran2,3, Sabiha Eddarkaoui4, Séverine Bégard4, Catherine Pythoud2,3,
Maria Rey2,3, Raphaëlle Caillierez4, Kim Q Do 1, Sophie Halliez 4, Paola Bezzi 5, Luc Buée 4,
Geneviève Leuba1, Morvane Colin4, Nicolas Toni 1,6 ✉ and Nicole Déglon 2,3,6

Alzheimer’s disease (AD) is characterized by the accumulation of the tau protein in neurons, neurodegeneration and memory
loss. However, the role of non-neuronal cells in this chain of events remains unclear. In the present study, we found accumulation
of tau in hilar astrocytes of the dentate gyrus of individuals with AD. In mice, the overexpression of 3R tau specifically in hilar
astrocytes of the dentate gyrus altered mitochondrial dynamics and function. In turn, these changes led to a reduction of adult
neurogenesis, parvalbumin-expressing neurons, inhibitory synapses and hilar gamma oscillations, which were accompanied by
impaired spatial memory performances. Together, these results indicate that the loss of tau homeostasis in hilar astrocytes of
the dentate gyrus is sufficient to induce AD-like symptoms, through the impairment of the neuronal network. These results are
important for our understanding of disease mechanisms and underline the crucial role of astrocytes in hippocampal function.

T
au is a microtubule-associated protein, abundant in the ner- pathological form of phospho-tau in different regions of the hippo-
vous system, which stabilizes microtubules and promotes campus in individuals with AD and in healthy age- and sex-matched
their assembly. Alternative splicing produces six tau isoforms donors (Supplementary Table 1 and Extended Data Fig. 1a–c). We
that can contain either three (3R) or four (4R) microtubule-binding used immunohistochemistry with an AD2 antibody, which recog-
repeats in the carboxy-terminal half, and between zero and two nizes the phosphorylated Ser396 and Ser404 epitopes7 (Fig. 1a). We
(0–2N) amino-terminal inserts; therefore, they are referred to as found strong variability in the density of AD2+ cells between hip-
0N3R, 1N3R, 2N3R, 0N4R, 1N4R and 2N4R1. In the healthy adult pocampal structures but also between individuals with AD, whereas
human brain, the 3R and 4R isoforms of tau are equimolar, but a healthy donors showed no AD2 immunoreactivity (Fig. 1b). For
disruption of the 3R to 4R ratio is sufficient to drive tau aggregation2 each individual, we found that the granule cell layer (GCL) of the
and the production of neurofibrillary tangles in pathological aging3 dentate gyrus and CA1 and CA3 regions exhibited a higher density
and tauopathies4. of AD2+ cells than the hilus and molecular layer (ML) of the dentate
By virtue of its unique plasticity and its integrative properties, gyrus (Fig. 1b). However, when individuals were graded according
the hippocampus plays a fundamental role in memory formation. In to the Braak staging, we found a strong correlation between stage
AD, as well as in several tauopathies, the hippocampal formation is and the density of AD2+ cells in the dentate gyrus, including the
largely impacted by the accumulation of hyperphosphorylated tau, ML, GCL and hilus (Fig. 1c,d). A high variability in the number of
accompanied by a reduction in synapse number, decreased adult hippocampal amyloid plaques was also observed between individu-
neurogenesis and neurodegeneration5. However, the contribution als and stages. However, we found no correlation between Braak
of non-neuronal cell types, and in particular of astrocytes, to the stage and hippocampal plaque number (Extended Data Fig. 1d–f).
functional deficiency of the hippocampus is unclear. In physiologi- Thus, in the hippocampus, the dentate gyrus and in particular the
cal conditions, astrocytes contribute to neuronal function and plas- hilus, are increasingly affected by the progression of tau pathology
ticity by several modes of regulation6. Thus, alterations in astrocytic in AD.
function may contribute to a disease phenotype, but the extent of We next examined the presence of 3R and 4R tau isoforms
this contribution is currently unclear. in the hilus of healthy donors and individuals with AD using
isoform-specific antibodies (Extended Data Fig. 2). The density of
Results 3R tau inclusions in the hilus was higher in individuals with AD
Accumulation of 3R tau in hilar hippocampal astrocytes of indi- who showed the presence of hyperphosphorylated tau, and this
viduals with AD. We examined the density of cells expressing a increase was exacerbated in individuals who also exhibited amyloid

Department of Psychiatry, Center for Psychiatric Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne,
1

Switzerland. 2Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center (CRN), Lausanne University Hospital (CHUV) and
University of Lausanne, Lausanne, Switzerland. 3Department of Clinical Neuroscience (DNC), Laboratory of Neurotherapies and Neuromodulation,
Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland. 4Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille
Neuroscience & Cognition, Lille, France. 5Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland. 6These authors
contributed equally: Nicolas Toni, Nicole Déglon. ✉e-mail: kevin.richetin@chuv.ch; nicolas.toni@unil.ch

Nature NeuroscIence | www.nature.com/natureneuroscience


Articles NATuRE NEuRoScIEncE

a Control Braak I Braak II Braak III Braak IV Braak V


CA3
ML CA3
CA1 CA1 GCL
Hilus
Hilus ML ML
ML
ML Hilus
+

GCL GCL
AD2

GCL
GCL Hilus
Hilus CA1
ML CA3
CA3
CA3

b 5,000
c ML GCL Hilus CA3 CA1
Control 2,000 5,000 3,000 4,000 3,000

2
AD2 cells/mm
P = 0.002 AD 1,500 4,000 3,000
3,000 2,000 2,000
4,000 P = 0.01 1,000 2,000
P = 0.0001 2,000 1,000 1,000
500 1,000
2

1,000

+
AD2 cells/mm

3,000 0 0 0 0 0
P = 0.004 0 2 4 6 0 2 4 6 0 2 4 6 0 2 4 6
0 2 4 6
2,000 P = 0.008 Braak stage Braak stage Braak stage Braak stage Braak stage
+

1,000 d Braak vs. ML Braak vs. GCL Braak vs. hilus Braak vs. CA3 Braak vs. CA1

R2 squared 0.1459 0.4313 0.5189 0.2571 0.1308


0
ML GCL Hilus CA1 CA3 P value 0.0655 0.0005 0.0001 0.019 0.0824

e f g h Control AD
Control AD

3R tau 4R tau NS

Control
– – – – P = 0.04
AD (P-tau /Aβ ) 1,000 P = 0.001 Control AD (P-tau /Aβ )
600 P = 0.02
P = 0.001
P = 0.04
800
2
inclusions/mm
+

600 400
Hilar 3R tau

2
inclusions/mm
+
Hilar 4R tau
+ – + +
+ –
AD (P-tau /Aβ )
+ +
AD (P-tau /Aβ ) 400 AD(P(P-tau
AD +/A) β -)
- Tau/Aβ AD (P-tau /Aβ )

200
200

0
0
P-tau – – + +
P-tau – – + +
Aβ – – – + Aβ – – – +

Fig. 1 | Accumulation of tau isoforms in the hilus of individuals with AD. a, Photomicrographs of the human hippocampus showing the density of
hyperphosphorylated tau (AD2+) cells in healthy controls and AD donors. b, Histogram showing the density of AD2+ cells in different hippocampal regions
of control and AD donors. c, Correlations between the density of AD2+ cells and Braak stage for each hippocampal area. d, Table showing the correlation
and P values between the density of AD2+ cells and Braak stage for each hippocampal area. e, Confocal micrographs showing the inclusions of 3R tau
in the hilus of control and AD donors. P-tau, hyperphosphorylated tau; Aβ, amyloid-β. f, Histogram showing the density of 3R tau inclusions in the hilus
of control and AD donors who were categorized according to the presence (+) or absence (−) of P-tau and Aβ. g, Confocal micrographs showing the
inclusions of 4R tau in the hilus of control and AD donors. h, Histogram showing the density of 4R tau inclusions in the hilus of control and AD donors who
were categorized according to the presence (+) or absence (−) of P-tau and Aβ. N = individuals/sections per individual: N = 9/4 for control, N = 21/4 for
AD; (b−d); N = 9/4 for control, N = 6/4 for AD (P-Tau−Aβ−); N = 6/4 for AD (P-Tau+Aβ−), n = 8/4 for AD (P-Tau+Aβ+); (f and h). Data are presented as the
mean ± s.e.m. Significance was determined by Mann–Whitney two-tailed t-test (b), one-sided ANOVA with two-tailed Tukey’s post hoc test (f and h) and
two-tailed Spearman’s rank non-parametric correlation test (d). Scale bars: 200 µm (a); 50 µm (e and g). NS, not significant.

plaques in the hilus. In contrast, the density of hilar 4R inclusions we found a high variability of 3R and 4R tau accumulation, which
was only increased in individuals devoid of hyperphosphorylated was not associated with disease state (Extended Data Fig. 3a–d).
tau or amyloid plaques, suggesting a transient increase along the The increased accumulation of 3R tau in astrocytes in the dis-
course of the disease (Fig. 1e–h). ease state was not due to modifications of S100β expression, since
AD is considered to be primarily a neuronal disease. However, the density of S100β+ cells was similar between individuals with AD
tau has also been found in astrocytes of individuals with AD, with and control donors (Extended Data Fig. 3e–h). Thus, disease state is
less well-known consequences8. We therefore examined the pres- associated with 3R tau accumulation in hilar astrocytes.
ence of 3R or 4R tau in astrocytes in the hilus of individuals with Synaptic failure is a major hallmark of AD resulting in a decrease9
AD. We observed more 3R tau inclusions per astrocyte and more or an increase in the density of synaptic proteins10, depending on
astrocytes with 3R tau inclusions in individuals with AD than in disease state and reactive mechanisms. We therefore assessed the
controls. Furthermore, this accumulation was greater in individuals expression of the presynaptic protein synaptophysin and the post-
with the presence of hyperphosphorylated tau and was exacerbated synaptic protein PSD95 using immunohistochemistry. The density
when the hilus exhibited amyloid plaques (Fig. 2a–d). In contrast, of PSD95 (Fig. 2h,i) but not of synaptophysin (Extended Data Fig.
no change in astrocytic accumulation of 4R tau was found with dis- 4) was significantly increased in the hilus of individuals with AD
ease state (Fig. 2e–g). In the non-astrocytic (S100β−) compartment, who showed the presence of hyperphosphorylated tau, an effect that

Nature NeuroscIence | www.nature.com/natureneuroscience


NATuRE NEuRoScIEncE Articles
was exacerbated by the presence of amyloid plaques. The density dorsal hippocampus, approximately 500 hilar cells per hippocam-
of PSD95 immunostaining strongly correlated with the propor- pus were infected (Fig. 3j). All transduced cells were found in the
tion of astrocytes accumulating 3R but not 4R tau, suggesting a link dentate gyrus, with a majority (65%) in the hilus. Similarly to the
between 3R tau accumulation in astrocytes and synaptic alterations LV-G1-GFP reporter construct, virtually all transduced cells were
(Fig. 2j,k). Thus, in AD, the hilus of the dentate gyrus is particularly identified as astrocytes (Fig. 3k), representing half of the astrocyte
responsive to disease progression and astrocytes accumulate 3R tau, population in the dorsal hilus (Fig. 3l). Astrocytes that accumulated
which is associated with synaptic alterations and the severity of the human 1N3R or 1N4R tau exhibited a pathological conformation of
pathology. tau, as assessed by immunohistochemistry using the MC-1 antibody
(Extended Data Fig. 6a).
Viral strategy for expressing tau in hilar astrocytes of adult mice. Thus, this LV enables the long-term expression of the human
Our observations from human samples raise the possibility that 3R tau isoforms in astrocytes of the dentate gyrus and in particular the
tau accumulation in hilar astrocytes may participate in hippocampal hilus, with very high anatomical and cellular specificity.
dysfunction and disease etiology. To test this possibility, we devel-
oped a new lentiviral vector (LV) to specifically target astrocytes. Tau isoform overexpression in astrocytes differentially affects
We used a truncated version of the glial fibrillary acidic protein mitochondrial distribution and function. By enabling their
(GFAP) promoter gfaABC1D, with a B3 enhancer (gfaABC1D(B3), coupling with the cytoskeleton, tau is known to play a role in
hereafter called G1) and the previously described miR124T neuro- organelle distribution and, in particular, mitochondria trans-
nal detargeting system (LV-G1-GFP; Fig. 3a). To assess the astro- port13. To assess the consequences of tau isoform accumulation
cytic specificity of this construct in the adult mouse brain, we on mitochondria, we used MitoTimer14. To examine the effect
injected the reporter LV-G1-GFP in the hilus of the dentate gyrus of 1N3R or 1N4R tau isoforms on mitochondria, we co-injected,
of adult mice. Two weeks after injection, we examined the distribu- into the mouse dentate gyrus, the following combinations of
tion and identity of the green fluorescent protein (GFP)-expressing LV: LV-G1-MitoTimer + LV-G1-CFP + LV-G1-1N3R-V5 or
cells. All GFP+ cells were found in the dentate gyrus, with a major- LV-G1-MitoTimer + LV-G1-CFP + LV-G1-1N4R-V5, or as control,
ity (68.4% ± 1.7%) in the hilus (Fig. 3b,c). Regardless of their posi- LV-G1-MitoTimer + LV-G1-CFP (or LV-G1-GFP). Four months
tion, virtually all GFP+ cells expressed GFAP (Fig. 3d) and S100β after injection, most astrocytes infected with the control construct
(Fig. 3e), which are astrocytic markers. Adult hippocampal stem (LV-G1-CFP) exhibited a uniform distribution of mitochondria
cells that reside in the subgranular zone of the dentate gyrus also in the soma, proximal processes (between 1 and 20 µm from the
express GFAP and could potentially be targeted by the LV. To assess soma) and distal processes (more than 20 µm from the soma),
stem cell targeting, we injected another set of mice. Four days after which was defined as a class 1-distribution pattern. A few astro-
LV injection, 8.1% ± 2.1% of GFP+ cells exhibited radial glial-like cytes (17.0% ± 9% of all control astrocytes) were devoid of mito-
(RGL) stem cell morphology, with a soma in the subgranular zone chondria in the distal processes (defined as a class 2 distribution
and a radial process extending into the GCL (Extended Data Fig. 5), pattern) or in distal and proximal processes (defined as a class 3
suggesting that few adult neural stem cells may have been targeted distribution pattern; 14.5% ± 2% of all control astrocytes; Fig. 4a,b).
by the LV. These cells represented 11% ± 1.2% of all RGL cells, In contrast, 1N3R tau overexpression strongly reduced the number
and this proportion decreased to 3.0% ± 1.1% at 14 and 120 days of mitochondria located in the distal processes, as evidenced by a
after LV injection. Furthermore, only 0.4% ± 0.4% of GFP+ cells significant increase in the proportion of class 3 and decrease in class
expressed the mature neuronal marker NeuN (Fig. 3f), suggesting 1 astrocytes (Fig. 4b and Extended Data Fig. 6b–d). In addition,
that the neuronal detargeting system disabled the transgene expres- 1N4R induced a redistribution of mitochondria towards the soma,
sion in new neurons as they matured. Together, these results show albeit less drastically than 1N3R (Fig. 4b). Mitochondrial relocation
that this approach enabled the specific targeting of hilar astrocytes towards the soma may be due to a retraction of astrocytic processes.
in the mouse hippocampus. To test this possibility, we analyzed the effect of 1N3R or 1N4R tau
We next used this strategy to express the human tau isoforms on astrocytic morphology. We found that 1N3R and 1N4R tau, were
in astrocytes. Since tau 0N is a fetal isoform and tau 2N is weakly homogeneously distributed throughout the soma and processes of
expressed in the human brain11, we used the 1N (1N3R and 1N4R) astrocytes (Fig. 4c). Furthermore, using GFP to examine astrocyte
isoforms linked to a V5 tag (LV-G1-1N3R or LV-G1-1N4R; Fig. 3g). morphology, we found that the projected area of individual astro-
We previously reported that the V5 tag does not interfere with tau cytic territories, the area of the soma, the number of branching
protein hyperphosphorylation and misfolding12 and it enables the points, the number of segments, the number of terminal points, the
detection of exogenous tau by western blot (Fig. 3h) and immu- total length of processes and neuropil infiltration volume (NIV; Fig.
nohistochemistry (Fig. 3i). Four months after the co-injection 4d,k) were similar between control and 1N3R or 1N4R astrocytes.
of LV-G1-GFP and either LV-G1-1N3R or LV-G1-1N4R in the Together, these results indicate that 1N3R, and to a lesser extent

Fig. 2 | 3R tau inclusions increase in the hilar astrocytes of individuals with AD. a, Low-magnification confocal micrographs of 3R (top) or 4R (bottom)
tau inclusions in hilar astrocytes (S100β, green; tau 3R and 4R, red) in control donors and donors with AD. b, Confocal micrographs and orthogonal
projections showing the presence of 3R tau inclusions in hilar astrocytes. c, Histogram of the density of 3R tau inclusions in hilar astrocytes of controls
or individuals with AD, who were distributed between 3 categories, depending on the presence of phosphorylated tau, amyloid plaques or both. d,
Histogram of the percentage of astrocytes that contained 3R tau inclusions in controls or individuals with AD. e, Confocal micrographs and orthogonal
projections showing the presence of 4R tau inclusions in hilar astrocytes. f, Histogram of the density of 4R tau inclusions in the hilar astrocytes of
controls or individuals with AD. g, Histogram of the percentage of hilar astrocytes that contained 4R tau inclusions in controls or individuals with AD. h,
Photomicrographs showing PSD95 immunostaining in the hilus of controls and the three categories of individuals with AD. i, Histogram of the intensity
of PSD95 staining in the hilus of controls or individuals with AD. j, Correlation between the intensity of PSD95 staining and the number of hilar astrocytes
expressing 3R tau. k, Correlation between the intensity of PSD95 staining and the number of hilar astrocytes expressing 4R tau. N = individuals/sections
per individuals/cells per section; N = 8/4/321 for control, N = 6/4/261 for AD (P-Tau−Aβ−), N = 6/4/258 for AD (P-Tau+Ab−), N = 7/4/287 for AD
(P-Tau+Ab+); (d and g). N = individuals/sections per individual; N = 7/4 for control, N = 6/4 for AD (P-Tau−Aβ−), N = 7/4 for AD (P-Tau+Ab−), N = 8/4 for
AD (P-Tau+Ab+); (i–k). Data are presented as the mean ± s.e.m. Significance was determined by one-sided ANOVA with Tukey’s post hoc test (c–f and i)
and two-tailed Spearman’s rank non-parametric correlation test (j–k). Scale bars: 250 µm (a and h); 5 µm (b and e).

Nature NeuroscIence | www.nature.com/natureneuroscience


Articles NATuRE NEuRoScIEncE

1N4R, induce a redistribution of mitochondria from the processes pocampal neuron–glial co-cultures infected with the
towards the soma without modifying the morphology of astrocytes. following combination of LV: LV-G1-MitoTimer + LV-G1-CFP
Next, we used the 555 nm/488 nm fluorescence ratio to exam- as control, or LV-G1-MitoTimer + LV-G1-CFP + LV-G1-1N3R or
ine the turnover and redox state of individual mitochondria. Both LV-G1-MitoTimer + LV-G1-CFP + LV-G1-1N4R. Similarly to our
1N3R and 1N4R tau increased the 555 nm/488 nm fluorescence in vivo observations, the LV targeted almost exclusively astrocytes,
ratio of mitochondria (Fig. 4l,m), indicating a reduced turnover and and most astrocytes were co-infected (Extended Data Fig. 7a–d).
increased oxidized state of mitochondria. Likewise, we found that mitochondria in control conditions were
To further investigate the consequences of 1N3R and uniformly distributed between proximal (between 1 and 20 µm from
1N4R tau isoform overexpression in astrocytes on mito- the soma) and distal processes (more than 20 µm from the soma)
chondrial dynamics and function, we used rat hip- of astrocytes (Fig. 5a–c). In contrast, 1N3R tau induced a drastic

a Control AD (P-tau /Aβ )


– – +
AD (P-tau /Aβ )

AD (P-tau /Aβ )
+ +
S100β/3R tau
S100β/4R tau

b c d AD
Control
400 P = 0.0009 100
P = 0.0001
80

Astrocytes with 3R
tau inclusions (%)
300 P = 0.0001
S100β/3R tau

+
2
S100β - 3R tau
inclusions/mm

60
200
+

40
100

+
20

0 0
P-tau – – + + P-tau – – + +
Aβ – – – + Aβ – – – +

e f g

200 80
S100β/4R tau

Astrocytes with 4R
tau inclusions (%)

150 60
+
2
S100β - 4R tau
inclusions/mm

100 40
+

50 20
+

0 0
P-tau – – + + P-tau – – + +
Aβ – – – + Aβ – – – +

h PSD95 in hilus i j k
– –
AD (P-tau /Aβ ) Control AD
Control
3R tau 4R tau
2
125 P = 0.0001 R = 0.25, P = 0.003 100
2
R = 0.0007, P = 0.91
100

P = 0.0036 80
100
PSD95 optical density

80
PSD95 optical density
PSD95 optical density

60 60
75

40 40
+ – + +
AD (P-tau /Aβ ) AD (P-tau /Aβ ) 50

20 20
25
0 0
0 20 40 60 80 100 0 20 40 60 80
0
+ +
P-tau – – + + +
S100β - 3R tau
+ S100β - 4R tau
+
+
/S100β (% of total) /S100β (% of total)
Aβ – – – +

Nature NeuroscIence | www.nature.com/natureneuroscience


NATuRE NEuRoScIEncE Articles
a Astrocyte promoter b DAPI/GFI c
gfaABC1D(B3) A B C1 D B B B BP Cortex 100 LV-G1-GFP

80
LV-G1-GFP LTR G1 GFP miR124T WPRE LTR

GFP cells (%)


CA1
Neuronal detargeting 60
system
40

+
C57BL/6
20

gy
Adult mouse CA3 Hilus GCL

lo
to
ML

is
0

H
ML GCL Hilus CA3 CA1
14 d.p.i.

d DAPI/GFP/GFAP e DAPI/GFP/S100β f DAPI/GFP/NeuN


125 125 125

100 100 GFP 100

GFP cells (%)


/GFP cells (%)

/GFP cells (%)

GFP NeuN /
+

+
GFP - GFAP

GFP - S100β

+
75 75 75

50 50 50

+
+
+
+

+
+
25 25 25

0 0 0

L
L
s
L
L
s

C
ilu

M
L
L
s
M
ilu

C
M
ilu

G
H
G

G
H

H
LV-G1-GFP
g i j LV-G1-1N3R
LV-G1-GFP LTR G1 GFP miR124T WPRE LTR LV-G1-1N4R
DAPI/GFP 600
LV-G1-1N3R LTR G1 1N3R miR124T WPRE LTR

2
Infected cells/µm
600
LV-G1-1N4R LTR G1 1N4R miR124T WPRE LTR
LV-G1-GFP

400
V5 tag 200

0
C57BL/6
Adult mouse k
gy

Astrocyte
lo
to
is

125 RGL cell


H

DAPI/1N3R-V5
Neuron
120 d.p.i.
100

Infected cells
(% of total)
LV-G1-1N3R

75
h 50
LV-G1-1N3R

LV-G1-1N4R

LV-G1-1N3R

LV-G1-1N4R
LV-G1-GFP

LV-G1-GFP

k
25

0
60 kDa

3R

4R
FP
1N

1N
G
1-
DAPI/1N4R-V5

1-

1-
-G
-G

-G
50 kDa

LV
LV

LV
Tau-V5

40 kDa l
LV-G1-1N4R

80

(% of total astrocytes)
30 kDa Infected cells
60

40
20 kDa
20
Hippocampus Cortex
0

Fig. 3 | Viral strategy to specifically target hilar astrocytes. a, Schematic of the vesicular stomatitis virus G (VSV-G) protein pseudotyped LV expressing
GFP and experimental timeline. LTR, long terminal repeat; WPRE, woodchuck post-regulatory element; d.p.i., days post injection. b, Confocal micrograph of a
mouse hippocampus injected with LV-G1-GFP (GFP, green; DAPI, blue), 14 d after LV injection. Inset: a GFP+ astrocyte. c, Histogram showing the distribution
of infected (GFP+) cells. d–f, Confocal micrographs and histograms of the distribution of cells that coexpressed GFP with GFAP (d), S100β (e) or NeuN (f). g,
Schematic of the VSV-G pseudotyped LV and experimental timeline. h, Western blots of hippocampal or cortical punches of animals injected with LV-G1-GFP,
LV-G1-1N3R-V5 or LV-G1-1N4R-V5, probed with anti-V5 antibody. i, Confocal micrographs of the hippocampus of mice injected with LV-G1-GFP, LV-G1-1N3R
or LV-G1-1N4 (DAPI, blue; GFP, green; V5, red). j, Histogram of the density of infected cells in the hilus of injected mice for the GFP (white), 1N3R (yellow)
or 1N4R (blue) constructs. k, Histogram of the percentage of infected cells with an astrocyte (orange), RGL cell (magenta) or neuron (green) phenotype.
l, Histogram of percentage of all S100β+ astrocytes in the hilus of mice that were targeted by each viral construct. Hil, hilus. N = animals/cells per animal;
LV-G1-CFP: 4/100–350 (c), LV-G1-1N3R: 4/100–350 (c and j), LV-G1-CFP: 4/50 (d–f and k–l), LV-G1-1N3R: 4/50 (d–f and k–l) and LV-G1-1N4R: 4/50 (d–f
and k–l). Data are presented as the mean ± s.e.m. Scale bars: 200 µm (b and i), 20 µm (b (inset) and d–f). Immunoblots (h) are cropped; full gel pictures are
shown in Supplementary Fig. 1.

relocation of mitochondria in proximal processes, whereas 1N4R ity, we used confocal live imaging to track mitochondrial move-
did not change the distribution of mitochondria in astrocytes (Fig. ment and found that 1N3R but not 1N4R increased the proportion
5c). Here too, tau isoforms did not induce morphological changes of of stationary mitochondria (Fig. 5d and Supplementary Videos 1
astrocytes (Extended Data Fig. 7e–k). and 2). Furthermore, by observing the movement of mitochondria
The effect of 1N3R tau on mitochondrial distribution may be relative to the soma, we found that 1N3R but not 1N4R reduced the
due to an effect on motility and dynamics. To assess this possibil- anterograde movement and increased the retrograde movement of

Nature NeuroscIence | www.nature.com/natureneuroscience


Articles NATuRE NEuRoScIEncE

a b P = 0.0054 Mitochondrial distribution


Class 1 Class 2 Class 3
P = 0.0001
LV-G1-CFP
100 P = 0.0001
LV-G1-1N3R P = 0.0001
(488 nm + 555 nm)

LV-G1-1N4R
80

Astrocytes (% of total)
60 P = 0.0001
CFP/MitoTimer

40

20

0
Class 1 Class 2 Class 3

c DAPI/GFP DAPI/V5 3D reconstruction d e f


LV-G1-CFP
4,000 500 500 LV-G1-1N3R
LV-G1-1N4R
LV-G1-GFP

400 400

Territory area (µm )


3,500

Number of nodes
Soma area (µm )
2
300 300
2,000
200 200

1,000
100 100

0 0 0
LV-G1-1N3R

g 1,000 h 800 i 5,000

Number of terminal points


800 4,000
Number of segments

600

Total processes
length (µm)
600 3,000
400
400 2,000
LV-G1-1N4R

200
200 1,000

0 0 0

j LV-G1-GFP LV-G1-1N3R LV-G1-1N4R k

600

#2 #1
#2 #2
DAPI/GFP/V5/NIV surface

#1
#1
NIV (µm ) 400
3

#3
#3 #3

200

l LV-G1-CFP LV-G1-1N3R LV-G1-1N4R m


50
Mitochondrial ratio (555 nm/488 nm)

40 LV-G1-CFP
Astrocytes (% of total)

LV-G1-1N3R
LV-G1-MitoTimer

P = 0.001
30 LV-G1-1N4R

20

10

0
0 0.2 0.4 0.6 0.8 1 1.2 1.4 1.6 1.8 2 2.2 2.4 2.6 2.8 3
Reduced Equilibrated Oxidized

Fig. 4 | Tau isoforms differentially affect mitochondria in hilar astrocytes in vivo. a, Representative confocal images of astrocytes displaying the three
classes of mitochondria distribution. b, Histogram of the distribution of hilar astrocytes in each class after infection with LV-G1-1N3R, LV-G1-1N4R or
control LV. c, Confocal micrographs of hilar astrocytes expressing GFP (left) and tau isoforms (red; middle) and 3D reconstructions of their soma and
processes (right). d–i, Morphological quantification of hilar astrocytes transduced with either LV-G1-GFP (white bars), LV-G1-1N3R (yellow) or LV-G1-
1N4R (blue) constructs, showing the projected territory area (d), surface of the soma (e), number of nodes (f), number of segments (g), number
of terminal points (h) and total length of processes (i). j, Confocal micrographs of hilar astrocytes expressing GFP (green) and tau isoforms (red;
insets) and representative NIVs after 3D reconstructions (magenta). k, Histogram showing the volume occupied by NIVs of astrocytes. l, Confocal
micrographs of astrocytes expressing MitoTimer (green and red), after infection with LV-G1-CFP + LV-G1-MitoTimer or LV-G1-1N3R (or LV-G1-
1N4R) + LV-G1-CFP + LV-G1-MitoTimer. m, Histogram of the mitochondrial redox state of hilar astrocytes after transduction. N = cultures/cells per culture;
LV-G1-CFP: 5/101, LV-G1-1N3R: 5/106, LV-G1-1N4R: 5/98; (b). LV-G1-CFP: 4/44, LV-G1-1N3R: 4/44, LV-G1-1N4R: 4/43; (d–i, k). LV-G1-CFP: 4/205, LV-G1-
1N3R: 4/196, LV-G1-1N4R: 5/249; (m). Data are presented as the mean ± s.e.m. Significance was determined by one-sided ANOVA with Tukey’s post hoc
test. Scale bars: 20 µm (a and l) and 15 µm (c), 10 µm (j) and 3 µm (j, inset).

Nature NeuroscIence | www.nature.com/natureneuroscience


NATuRE NEuRoScIEncE Articles
mitochondria (Fig. 5e). Thus, 1N3R tau reduced the total motility mitochondria. In contrast, 1N4R showed mild effects on mitochon-
of mitochondria and induced their transfer to the soma, resulting drial localization and did not impair mitochondrial function. We,
in a decrease in mitochondria in the distal segments of astrocytic therefore, next focused on the functional implications of 1N3R
processes. overexpression.
Movement enables the recycling of damaged mitochondria,
their fusion and fission, as well as biogenesis, all of which sustain 1N3R tau overexpression in hilar astrocytes impairs the hippo-
mitochondrial function15. We therefore expected the scarce, sta- campal neuronal network. To assess the long-term consequences
tionary, distal mitochondria to exhibit morphological and func- of 1N3R tau accumulation in hilar astrocytes on neurons, we
tional impairment. We first examined the morphology of individual injected LV-G1-CFP (control) or LV-G1-1N3R + LV-G1-CFP into
mitochondria. As compared to control conditions, 1N3R but not the hilus of adult mice. Four months later, we evaluated the den-
1N4R tau overexpression in astrocytes reduced mitochondrial pro- sity of different neuronal populations in the hilus. In both condi-
jected area, especially in the distal processes (Fig. 5f), suggesting tions, we observed a similar density of cells (DAPI+; Fig. 6a) and
that the mitochondria that remained in the distal processes may be of neurons (NeuN+; Fig. 6b) in the hilus, suggesting that 1N3R tau
impaired. Next, we examined the redox state/turnover of mitochon- overexpression in astrocytes did not induce cell death. In contrast,
dria using the using the MitoTimer reporter gene. Neither 1N3R 1N3R tau overexpression in astrocytes significantly decreased the
nor 1N4R altered the turnover/redox state of mitochondria in the number of neurons expressing the activity-dependent protein par-
soma. However, both isoforms increased the redox state in the mito- valbumin (PV, Fig. 6c), whereas the density of mossy cells was not
chondria that had remained in the proximal and distal processes changed (GluR2/3+; Fig. 6d). This suggests that 1N3R tau overex-
(Fig. 5g,h). Thus, the in vitro experiments confirmed and extended pression in astrocytes affected PV expression in interneurons and,
the in vivo observations showing that 1N3R tau overexpression in consequently, reduced the inhibitory transmission. The dentate
astrocytes induced mitochondrial relocation in the soma, concomi- gyrus is one of the two major sites for adult neurogenesis to occur,
tant with alterations in mitochondrial morphology and function in and a dysregulation of astrocytes or neuronal activity may interfere
the distal processes, whereas 1N4R had a modest impact, restricted with this process18. We quantified cell proliferation by injecting ani-
to mitochondrial motility and redox state. mals with the thymidine analog 5-bromo-2′-deoxyuridine (BrdU)
In astrocytes, mitochondrial dynamics and redox state/turn- and analyzing animals 1 d after injection. We found that the num-
over have an impact on intracellular ATP production and the ber of cells that incorporated BrdU in the subgranular zone was
regulation of intracellular calcium concentration16. To investigate unchanged in 1N3R tau-overexpressing animals (Fig. 6e), suggest-
the consequences of 1N3R and 1N4R tau overexpression in astro- ing that the few RGL cells that were targeted by the LV (Extended
cytes on mitochondrial ATP production, we used a Förster reso- Data Fig. 5) were not sufficient to influence cell proliferation in the
nance energy transfer (FRET)-based sensor for ATP production. dentate gyrus. To assess the later stages of adult neurogenesis, we
To this aim, we co-infected astrocyte–neuron co-cultures with used immunostaining against the cytoskeletal marker of immature
an LV encoding either the cyan fluorescent protein (LV-G1-CFP neurons, doublecortin (DCX). The number of DCX-expressing
as control) or LV-G1-1N3R or LV-G1-1N4R, together with a LV cells was significantly reduced in 1N3R tau-expressing animals (Fig.
encoding a FRET-based fluorescent mitochondrial ATP probe17 6f), suggesting an impaired maturation of newborn neurons.
(LV-G1-MitoGoAteam2). In astrocytes overexpressing 1N3R tau Next, we examined inhibitory synapses in the hilus. Inhibitory
but not in astrocytes overexpressing 1N4R tau, we found that ATP synapses are composed of nanoscale subsynaptic domains where
production by individual mitochondria was significantly reduced in the scaffolding protein gephyrin and the GABA vesicle transporter
distal processes as compared to control astrocytes (Fig. 5i,j). In con- VGAT are closely associated (within 300 nm of each other19). Using
trast, we found no difference in ATP production in mitochondria three-dimensional (3D) confocal reconstructions, we evaluated
of the soma or proximal processes (Fig. 5j). Finally, we investigated the density of VGAT and gephyrin clusters in the territories of
the calcium concentration in the soma and proximal and distal pro- 1N3R-overexpressing astrocytes (Fig. 6g,h). As compared to control
cesses using the Fluo-4 AM calcium sensor. Compared to control, mice, we observed a decrease in gephyrin dots, resulting in a reduc-
1N3R tau overexpression had no impact on calcium concentration tion of paired VGAT–gephyrin punctae, which indicates a reduc-
in the soma and proximal processes but significantly decreased tion of inhibitory synapses (Fig. 6i–l). Thus, the overexpression
calcium concentrations in distal processes, whereas 1N4R did not of 1N3R tau in astrocytes impaired inhibitory neurons and adult
influence calcium concentrations (Fig. 5k,l). Overall, these results neurogenesis.
show that in vitro, 1N3R tau overexpression in astrocytes induced Together, these results suggest that 1N3R tau overexpression in
the relocation of mitochondria from the distal processes to the soma astrocytes may impair the function of the neuronal network in the
and proximal processes, which resulted in few and impaired distal hilus of the dentate gyrus. To assess the basal activity of the hilus, we

Fig. 5 | Tau isoforms differentially alter mitochondrial function in astrocytes in vitro. a, Schematic of hippocampal neuron–glial co-cultures showing
the proximal (<20 µm from the soma) and distal (>20 µm) portions of astrocytic processes. b, Confocal micrographs of mitochondria (white)
in astrocytes (blue) 14 d after infection with LV-G1-CFP + LV-G1-MitoTimer (control) or LV-G1-1N3R + LV-G1-CFP + LV-G1-MitoTimer, or LV-G1-
1N4R + LV-G1-CFP + LV-G1-MitoTimer. Bottom: higher magnification views. c, Histogram of mitochondrial distribution in proximal and distal processes
after LV transduction. d, Histogram of the percentage of stationary, mobile and highly mobile mitochondria after LV transduction in the proximal (left) and
distal (right) processes. e, Histogram of the percentage of anterograde or retrograde mitochondrial motility. f, Histogram of the surface of mitochondria
in proximal and distal processes. g, Confocal micrographs of MitoTimer (green and red) in CFP+ (blue) astrocytes after expression of CFP alone or CFP
plus 1N3R or 1N4R tau. h, Histogram of the redox-state ratio in the soma, proximal and distal processes after LV transduction. i, Confocal micrographs of
MitoGoAteam2 (green and red) in CFP+ (blue) astrocytes after expression of CFP alone or CFP plus 1N3R or 1N4R tau. j, Histogram of the ATP level in
the soma, and proximal and distal processes of astrocytes. k, Confocal micrographs of Fluo-4 AM (red) in control CFP+ (blue) astrocytes and astrocytes
expressing 1N3R or 1N4R tau. l, Histograms of the estimated intracellular calcium concentrations in the soma and processes of astrocytes. N = cultures/
cells per culture; LV-G1-CFP: 5/127, LV-G1-1N3R: 6/152, LV-G1-1N4R: 6/154; (c). LV-G1-CFP: 4/20, LV-G1-1N3R: 4/19, LV-G1-1N4R: 4/23; (d–f). LV-G1-CFP:
4/23, LV-G1-1N3R: 4/21, LV-G1-1N4R: 4/23; (h, j, l). Data are presented as the mean ± s.e.m. Significance was determined by one-sided ANOVA with
Tukey’s post hoc test. Scale bars: 20 µm (b) and 2 µm (g, i and k).

Nature NeuroscIence | www.nature.com/natureneuroscience


Articles NATuRE NEuRoScIEncE

examined the expression of the immediate-early gene c-Fos. LV-G1- the generation of gamma oscillations, which enable coincidence
1N3R-injected animals showed fewer c-Fos+ cells in the hilus than detection and regulate circuit performance20. We therefore exam-
control mice, suggesting reduced neuronal activity in the hilus of ined evoked gamma oscillations in the dentate gyrus of acute hip-
these mice (Fig. 6m). PV-expressing interneurons are crucial for pocampal slices using extracellular electrophysiological recordings.

a b LV-CFP LV-1N3R-V5 LV-1N4R-V5 c


LV-G1-CFP
Neuron–glia hippocampal 150 LV-G1-1N3R
co-culture CFP/MitoTimer(488 nm + 555 nm) NS LV-G1-1N4R

Mitochondrial processes
P = 0.0001 P = 0.02 NS
100

distribution
P = 0.0001 P = 0.02

Soma
50
Proximal (<20 µm)
Distal (>21 µm)
0
Proximal Distal

d Proximal Distal e f
125 25
Highly mobile 100
Mitochondria (% of total)

P = 0.0042 P = 0.0042 P = 0.016


100 Mobile

Mitochondrial surface (µm2)


80 25
Motility (% of total)

Stationary
75
60 20
50
P = 0.0001

P = 0.0001

40 15
25

Retro
Antero
20 10
0
4R

4R
P

1- R

1- R
P

5
-G CF

0
-G CF
-G N3

3
1N

1N
LV -1N

Antero Retro
1-

LV 1-1

LV 1-
-G

1
-G

-G
LV

LV

0
LV

g Soma Proximal Distal h


P = 0.015

LV-G1-CFP
P = 0.003
6
Mitochondrial redox state

P = 0.016
CFP/MitoTimer

LV-G1-1N3R 4
P = 0.016

2
LV-G1-1N4R

0
Soma Proximal Distal

i j
LV-G1-CFP
3
Mitochondrial ATP level
CFP/MtoGoAteam2

LV-G1-1N3R 2 P = 0.007

1
LV-G1-1N4R

0
Soma Proximal Distal

k l
LV-G1-CFP 2.5 P = 0.0002

2.0
CFP/Fluo-4 AM

LV-G1-1N3R 1.5
(Ca2+)i

1.0

LV-G1-1N4R 0.5

0
Soma Proximal Distal

Nature NeuroscIence | www.nature.com/natureneuroscience


NATuRE NEuRoScIEncE Articles
Transient high-frequency oscillatory activity was induced by a brief tion of PV interneurons observed after 1N3R tau overexpression
focal application of glutamate in the hilus (Fig. 6n–t)21. We observed in astrocytes contributes to the behavioral impairment observed in
two distinct types of oscillatory activity: a gamma oscillatory activ- these mice. To test this possibility, we used the neuregulin 1 pep-
ity (mean peak frequency ~75 Hz) which relies on functional tide (NRG1p) to increase PV interneuron excitability26. Another
GABAA receptor transmission (Fig. 6o–q)21,22 and a faster oscillatory set of animals was injected with LV-G1-1N3R or LV-G1-GFP. Four
activity (mean peak frequency ~100 Hz), similar to the nonsynaptic months later, mice were injected with NRG1p or vehicle and, 1 h
network synchrony described by Towers et al.22 (Fig. 6r–t). In slices later, tested on the new object location test. Similarly to the cohort
obtained from mice injected with the LV-G1-1N3R vector, we found of mice shown in Fig. 7d, 1N3R tau-vehicle injected mice showed
that the power of gamma oscillations (within the range of 50 to a lack of preference for the displaced object, as compared to con-
90 Hz) was significantly reduced as compared to slices from control trol groups. The recognition of the displaced object was however
mice (Fig. 6q). Likewise, the peak frequency of these gamma oscil- restored in LV-G1-1N3R animals injected with NRG1p (Fig. 7j),
lations was significantly lower in 1N3R tau mice (68.6 Hz ± 1.6 Hz; suggesting that increasing PV interneuron activity restored the
mean ± s.e.m.) than in control mice (75.1 Hz ± 1.3 Hz, P < 0.01; effect of 1N3R tau expression in astrocytes. To assess the involve-
Fig. 6p). In contrast, we observed no significant difference in the ment of PV interneurons in this effect, we examined PV immunore-
power and the peak frequency of the faster oscillatory activity (Fig. activity immediately after the behavioral test. LV-G1-1N3R-injected
6s). Thus, the accumulation of human 1N3R tau in hilar astrocytes mice showed a reduced number of PV+ cells and a reduced density
impaired synchronous activity. of PV immunoreactivity in the hilus as compared to control groups,
and the density of PV immunoreactivity was restored to control val-
Hilar astrocytic 1N3R tau accumulation affects spatial memory ues upon NRG1p injection (Fig. 7k–n).
of adult mice. Gamma oscillations in the dentate gyrus play a role Thus, 1N3R tau expression in hilar astrocytes reduced long-term,
in spatial memory23 and are impaired in mouse models of AD24. spatial memory performances, which were restored upon stimula-
We therefore compared the hippocampal-dependent spatial mem- tion of PV interneurons by NRG1p injection.
ory performances of mice 4 months after the bilateral injection of
LV-G1-CFP as control or LV-G1-1N3R LV (Fig. 7a). We first used Discussion
the new object location task and found that the preference index In individuals with AD, we found that astrocytes of the hilus of the
for the displaced object was significantly higher than chance level dentate gyrus accumulate 3R but not 4R tau, and this accumulation
for the control but not for the LV-G1-1N3R group, indicating a is correlated with synaptic alterations, suggesting an important role
reduction of spatial memory (Fig. 7b−d). On another set of mice, for astrocytes in disease progression. Using a new LV to specifically
we performed the Morris water maze test. LV-G1-1N3R-injected target astrocytes of the hilus, we found that overexpression of the
mice showed similar performances to control mice in the learning human 1N3R isoform of tau in these cells strongly impaired mito-
phase and the probe test (Fig. 7e–g). However, they showed a deficit chondrial motility, distribution and function, resulting in impaired
in reversal memory (Fig. 7h,i), indicating a difficulty to suppress neurogenesis, reduced number of neurons expressing PV, decreased
old spatial memory. To assess whether the decreased performances density of inhibitory synapses and reduced gamma oscillatory activ-
were specific to spatial memory, four independent behavioral tests, ity. Together, these modifications led to impaired spatial memory,
unrelated to spatial memory, were performed on the same sets of which was restored by stimulating PV interneuron activity.
mice: dark/light box test, which assesses anxiety; Y-maze (YM) Although tau has been found in glial cells27, astrocytes do not
for spatial working memory; object recognition task for nonspa- express this protein in physiological conditions28, and the origin of
tial long-term memory; and contextual fear conditioning, which tau in astrocytes in AD is unclear. One unsubstantiated possibility is
assesses fear memory. For all these tests, LV-G1-1N3R-injected that AD progression induces tau translation from the mRNA pres-
mice performed similarly to control mice (Extended Data Fig. 8). ent in astrocytes29. Alternatively, astrocytes may capture extracel-
Thus, the long-term overexpression of 1N3R tau in hilar astrocytes lular tau. Indeed, tau is released in the interstitial fluid by neurons30,
is sufficient to specifically alter spatial memory. spreads between cells31 and astrocytes can uptake tau when exposed
PV interneurons play an important role in hippocampal function to this protein32. Furthermore, tau was found in extracellular ves-
and spatial memory25. However, it is unclear whether the reduc- icles from the cerebrospinal fluid of individuals with AD33, which

Fig. 6 | Impact of astrocytic tau overexpression on hippocampal function. a–f, Immunofluorescence and confocal microscopy micrographs (left) and
evaluations (right) of cell populations, 4 months after LV injection with LV-G1-GFP alone (white bars) and LV-G1-GFP + LV-G1-1N3R (yellow bars). a,
DAPI+ cells (blue) in the hilus. b, NeuN+ neurons in the dentate gyrus. c, PV+ neurons (red) in the hilus. d, GluR2/3+ neurons in the hilus. e, BrdU+ cells in
the subgranular zone of the dentate gyrus. f, DCX+ cells in the GCL of the dentate gyrus. g, Confocal micrograph (top) and 3D reconstruction (bottom),
showing distal processes of astrocytes (green), in proximity of gephyrin (blue) and VGAT (red) clusters. h, Schematic (top), 3D reconstructions (middle)
and confocal micrographs (bottom) of VGAT and gephyrin clusters, unpaired (left) or paired (middle and right). i–l, Quantification of VGAT and gephyrin
clusters in the territories of hilar astrocytes transduced with either a LV-G1-GFP (control) or a LV-G1-1N3R construct. i, Density of VGAT+ clusters. j,
Density of paired VGAT–gephyrin clusters. k, Density of gephyrin clusters. l, Gephyrin:VGAT cluster ratio. m, Confocal micrographs and quantification of
c-Fos expression. n, Schematic of the approximate position of the extracellular electrodes and local glutamate injection for oscillatory activity analysis. o,
Time–frequency plots of glutamate-induced gamma oscillatory activity (recordings with a peak frequency between 50 and 80 Hz) in the dentate gyrus.
P values (right) represent the statistical difference between groups. p, Power spectra (mean ± s.e.m.) of glutamate-induced gamma oscillatory activity.
q, Power of gamma oscillations measured between 50 and 90 Hz. r, Time–frequency plots of glutamate-induced fast oscillatory activity (recordings
with a peak frequency between 90 and 110 Hz). P values (right) represent the statistical difference between groups. Time 0 corresponds to the onset
of glutamate injection. s, Power spectra of glutamate-induced fast oscillatory activity. t, Power of fast oscillations measured between 80 and 120 Hz.
N = animals/sections per animal; LV-G1-CFP: 6/5, LV-G1-1N3R: 6/5; (a–f). N = animals/cells per animal; LV-G1-CFP: 4/21, LV-G1-1N3R: 4/24; (i–l).
N = animals/sections per animal; LV-G1-CFP: 6/5, LV-G1-1N3R: 6/5; (m). N = 6 animals per group and 10 cells per animal (i–l). N = 6 animals per group and
n = 35–40 recordings (n–p). N = 6 animals per group and n = 16–20 recordings (q–s). Data are presented as the mean ± s.e.m. Significance was determined
by Mann–Whitney two-tailed t-test (a–f, i–l, q and t) and one-sided ANOVA with two-tailed Tukey’s post hoc test (p and s). Scale bars: 20 µm (a–f and
m), 5 µm (g) and 250 nm (h).

Nature NeuroscIence | www.nature.com/natureneuroscience


Articles NATuRE NEuRoScIEncE

may also contribute to the intercellular propagation of this protein34. In tauopathies, much attention has been given to the role of tau
These possibilities are currently under intense scrutiny. in neurons35. However, in many tauopathies, tau is found in glial
cells36, with poorly known consequences for disease symptoms and

a LV-GFP LV-1N3R-V5 d LV-GFP LV-1N3R-V5


10,000 800 LV-G1-CFP
LV-G1-1N3R
8,000
600

DAPI/GFP/Glur2/3
2

2
6,000

Cells/mm

Cells/mm
DAPI/GFP

400
4,000

200
2,000

0 0
+ +
DAPI GluR2/3
b e 2,500
4,000

2,000
3,000
DAPI/GFP/NeuN

DAPI/GFP/BrdU

2
1,500

Cells/mm
2
Cells/mm

2,000
1,000

1,000 500

0 0
+
NeuN
+ BrdU
c f 10,000
P = 0.0001
800 P = 0.0016

8,000
DAPI/GFP/DCX
600
DAPI/GFP/PV

2
6,000

Cells/mm
2
Cells/mm

400
4,000

200 2,000

0
0 +
+ DCX
PV

g h i j P = 0.0015
60 15
Unpaired Paired
(>300 nm) (<300 nm)

Paired VGAT-gephyrin
3
VGAT dots/100 µm

40 10
VGAT

3
dots/µm
20 5
Gephyrin 1:1 1:2 1:3
GFP/Gephyrin/VGAT

0 0

k l P = 0.0007
100 P = 0.0006 4
3

80
Gephyrin dots/100 µm

Gephyrin:VGAT ratio 3
60
2
40

1
20
250 nm
0 0

m DAPI/GFP/c-Fos Oscillatory activity ≤ 90 Hz)

o LV-G1-GFP LV-G1-1N3R dB P value p 250 q P = 0.014


14.5 1

120 25,000
200
Power (µV × Hz)

Gamma power oscillations

100 7.3
20,000
150
Frequency (Hz)

0.1
2

(50–90 Hz; µV )
2

80 15,000
0 100

60 0.01
50 10,000
–7.3
300 40
0 5,000
20 10 30 50 70 90 110 130 150
c-Fos cells/mm2

–14.5 0.001
P = 0.025
200 0 5 10 0 5 10 0 5 10 Frequency (Hz) 0
Time (s)
+

100
Oscillatory activity (≥ 90 Hz)
r LV-G1-GFP LV-G1-1N3R dB P value s t 25,000
150 16.8 1 300
0
20,000
130
Fast power oscillations

8.4
Power (µV * Hz)

n
(80–120 Hz; µV )

200
2
Frequency (Hz)

0.1

110 15,000
2

90 100 10,000
0.01
Local injection
DG of glutamate –8.4
(10 mM)
for 0.2 s 70 5,000
0
50 –16.8 0.001
10 30 50 70 90 110 130 150 0
0 5 10 0 5 10 0 5 10
Time (s) Frequency (Hz)

Nature NeuroscIence | www.nature.com/natureneuroscience


NATuRE NEuRoScIEncE Articles
a b c d P = 0.0009
Displaced object 80
LV-G1-CFP 70 ###
Spatial memory Stable object
LTR G1 CFP mir124T WPRE LTR

Preference Index for


displaced object(%)
60

Exploration time (s)


60
LV-G1-1N3R 50
LTR G1 1N3R mir124T WPRE LTR 40 40
Trial 24 h 30
Test 20 20

Behavior
C57bL/6
adult mouse 10
Object location 0 0

3R
FP
LV-G1-CFP

1N
C
4 months after LV-G1-1N3R

1-

1-
G

G
injection

-
LV

-
LV
e f g Probe 24 h h i
Spatial learning Reversal
P = 0.0001 LV-G1-GFP
100 P = 0.0001 100
100 P = 0.007 Reversal LV-G1-1N3R
80 morris water maze 80

Escape latency (s)


80
Escape latency (s)

Morris water maze P = 0.0012

Time in quadrant
(% of total)
60 60 60

40 40 40

20 20 20

0 0 0
1 2 3 4 5 6 Others Target 1 2 3
Training block Training block

k LV-G1-GFP + saline LV-G1-1N3R + saline LV-G1-1N3R + NRG1p l 2,000


P = 0.0091

1,500

PV+ cells/mm2
DAPI/GFP/PV

j
P = 0.0015 1,000
100
#
Preference Index for
displaced object (%)

80 ## 500

60
0
40
m n
P = 0.0001
20 60,000

PV labeling intensity (a.u.)


0
P = 0.0028
DAPI/GFP/PV

LV-G1-GFP + saline 40,000


LV-G1-1N3R + saline
LV-G1-1N3R + NRG1p
20,000

Fig. 7 | Tau 3R overexpression in hilar astrocytes induces a spatial memory deficit that is restored by NRG1p injection. a, Schematic of the VSV-G
pseudotyped LV and the experimental design used to evaluate the impact on cognitive function. Mice were injected with LV-G1-GFP alone or
LV-G1-GFP + LV-G1-1N3R. b, Schematic of the object location test. c, Histogram showing the time spent interacting with the immobile and displaced
objects. d, Histogram of the percentage of time spent interacting with the displaced object. e, Schematic of the learning task with Morris water maze. f,
Histogram showing the latency to find the hidden platform. g, Histogram showing the average number of crossings above the location of the target platform
1 d after spatial training. h, Schematic of the learning reversal task. i, Histogram of the latency to find the hidden platform. j, Histogram of the percentage
of time spent interacting with the displaced object of animals 1 h after intraperitoneal injection of either a saline solution (control) or NRG1p. k, Confocal
micrographs of hippocampal slices showing DAPI (blue), PV+ cells (red) and infected astrocytes (with LV-G1-GFP or LV-G1-GFP + LV-G1-1N3R) after
intraperitoneal injections with saline solution or NRG1p. l, Histogram of the density of PV+ neurons in the hilus. m, Confocal micrographs of PV+ neurons
in the hilus; DAPI (blue), PV (red) and infected astrocytes (green). n, Histogram of the optical density of PV immunostaining in the hilus; a.u., arbitrary
units. n = animals; LV-G1-CFP + saline: 8, LV-G1-1N3R + saline: 7, LV-G1-1N3R + NRG1p: 7; (j). LV-G1-CFP: 8, LV-G1-1N3R: 8 (c, d, f, g and i). n = animals/cells
per animal; LV-G1-CFP + saline: 6/144, LV-G1-1N3R + saline: 7/204, LV-G1-1N3R + saline: 4/68; (l and n). Significance was determined by Mann–Whitney
two-tailed t-test (c and d), Wilcoxon signed-rank test to chance level with ###P < 0.001, ##P < 0.05 and #P < 0.01 (d–j), one-sided ANOVA with two-tailed
Tukey’s post hoc test (j, l, n) and two-sided ANOVA with two-tailed Dunnett’s post hoc test (f, g, i). Data are presented as the mean ± s.e.m. Scale bars:
25 µm (k) and 10 µm (m).

progression. Disentangling the contribution of different cell types to Similarly, the contribution of small brain regions to specific func-
a given phenotype is crucial for our understanding of disease etiol- tions is difficult to assess without tools that selectively target them.
ogy. However, access to this information is often hampered by the In this study, we achieved the first goal by using a new LV strategy
lack of specific tools that selectively target subpopulations of cells. that enabled the expression of the genes of interest in astrocytes,

Nature NeuroscIence | www.nature.com/natureneuroscience


Articles NATuRE NEuRoScIEncE

with negligible expression in non-astrocytic cell types, both in vitro results show that their impairment can contribute to memory dis-
and in vivo. Upon injection into the hilus, the limited diffusion of turbances and may dramatically worsen disease symptoms.
the LV further enabled the exclusive targeting of the dentate gyrus,
since all transduced cells were found in the dentate gyrus and about Online content
70% in the hilus. This targeting, both at the anatomical and at the Any methods, additional references, Nature Research report-
cellular levels, enabled us to reproduce in mice, the observations we ing summaries, source data, extended data, supplementary infor-
obtained from human participants. mation, acknowledgements, peer review information; details of
Using this approach, we found that 1N3R and 1N4R tau overex- author contributions and competing interests; and statements of
pression in astrocytes differentially altered their mitochondrial local- data and code availability are available at https://doi.org/10.1038/
ization, trafficking and function, as well as calcium buffering. These s41593-020-00728-x.
effects may be mediated by several mechanisms: first, tau competes
with kinesin/dynein cargoes for microtubules, which are involved in Received: 3 September 2018; Accepted: 24 September 2020;
mitochondria transport in astrocytes. Interestingly, tau affinity for Published: xx xx xxxx
microtubules differs between the 3R and 4R isoforms37, a difference
that may underlie their differential effect on astrocytic mitochon- References
1. Buee, L. et al. Tau protein isoforms, phosphorylation and role in
dria. Furthermore, according to models of multiple-motor-driven neurodegenerative disorders. Brain Res. Rev. 33, 95–130 (2000).
cargo transport, the higher solubility and kinesin inhibitory activ- 2. Adams, S. J., de Ture, M. A., McBride, M., Dickson, D. W. & Petrucelli, L.
ity38 of 3R tau compared to 4R tau may induce a strong steric inhi- Three repeat isoforms of tau inhibit assembly of four repeat tau filaments.
bition of the binding strength of mitochondria to microtubules, PLoS ONE 5, e10810 (2010).
leading to their immobilization. Alternatively, the effect of tau on 3. Panda, D., Samuel, J. C., Massie, M., Feinstein, S. C. & Wilson, L. Differential
regulation of microtubule dynamics by three- and four-repeat tau:
mitochondrial transport may be due to posttranslational modifica- implications for the onset of neurodegenerative disease. Proc. Natl Acad. Sci.
tions, such as phosphorylation or truncation, which modulate tau USA 100, 9548–9553 (2003).
functions. Indeed, tau truncation produces N-terminal fragments, 4. Conrad, C. C. et al. Single-molecule profiling of tau gene expression in
which modulate kinesin velocity, and overexpression of which Alzheimer’s disease. J. Neurochem. 103, 1228–1236 (2007).
5. Hamilton, L. K. et al. Aberrant lipid metabolism in the forebrain niche
alters the mitochondrial system39. Furthermore, tau overexpression
suppresses adult neural stem cell proliferation in an animal model of
in astrocytes disrupts the intermediate filament network40, which Alzheimer’s disease. Cell Stem Cell 17, 397–411 (2015).
may impair the transport of other cargo, including peroxisomes and 6. Santello, M., Toni, N. & Volterra, A. Astrocyte function from information
endosomes41, disrupts the blood–brain barrier42, reduces the expres- processing to cognition and cognitive impairment. Nat. Neurosci. 22, 154–166
sion of glutamate transporters and reduces gliotransmitter release43. (2019).
7. Buée-Scherrer, V. et al. AD2, a phosphorylation-dependent monoclonal
Alone or in combination, these effects are consistent with our antibody directed against tau proteins found in Alzheimer’s disease. Mol.
observations of impaired mitochondrial transport and function. In Brain. Res. 39, 79–88 (1996).
turn, since astrocytes are involved in diverse brain functions44, the 8. Kovacs, GaborG. et al. Aging-related tau astrogliopathy (ARTAG):
impairment of astrocytic function is expected to impact on the neu- harmonized evaluation strategy. Acta Neuropathol. 2, 87–102 (2015).
ronal network and on behavior. 9. Bereczki, E. et al. Synaptic proteins predict cognitive decline in Alzheimer’s
disease and Lewy body dementia. Alzheimers Dement. 12, 1149–1158 (2016).
It is noteworthy that the impairment of a few hundred astrocytes 10. Savioz, A., Leuba, G. & Vallet, P. G. A framework to understand the
in the hilus of the dentate gyrus altered hippocampal function and variations of PSD-95 expression in brain aging and in Alzheimer’s disease.
spatial memory. Although our results do not rule out that other Ageing Res. Rev. 18, 86–94 (2015).
brain areas may display astrocytic tau accumulation in the course 11. Boutajangout, A., Boom, A., Leroy, K. & Brion, J. P. Expression of tau mRNA
of AD, the dentate gyrus is crucial for hippocampal function and and soluble tau isoforms in affected and non-affected brain areas in
Alzheimer’s disease. FEBS Lett. 576, 183–189 (2004).
memory performances45. In particular, our results point to two 12. Dujardin, S. et al. Neuron-to-neuron wild-type tau protein transfer through a
major effects of disrupted astrocytes on the function of the dentate trans-synaptic mechanism: relevance to sporadic tauopathies. Acta
gyrus: adult neurogenesis and PV interneuron function. Adult neu- Neuropathol. Commun. 2, 14 (2014).
rogenesis occurs in several steps, from stem/progenitor cell prolifer- 13. Cheng, Y. & Bai, F. The association of tau with mitochondrial dysfunction in
ation to the differentiation and maturation of new neurons, several Alzheimer’s disease. Front. Neurosci. 12, 2014–2019 (2018).
14. Gottlieb, R. A. & Stotland, A. MitoTimer: a novel protein for monitoring
of which are regulated by astrocytes46. Here, we found that 1N3R mitochondrial turnover in the heart. J. Mol. Med. 93, 271–278 (2015).
tau expression in astrocytes did not affect proliferation, but strongly 15. Eisner, V., Picard, M. & Hajnóczky, G. Mitochondrial dynamics in adaptive
reduced the number of immature neurons, similarly to recent and maladaptive cellular stress responses. Nat. Cell Biol. 20, 755–765 (2018).
observations in the human AD brain47. These results are consistent 16. Agarwal, A. et al. Transient opening of the mitochondrial permeability
with the role of gliotransmitters in the maturation of adult-born transition pore induces microdomain calcium transients in astrocyte
processes. Neuron 93, 587–605 (2017).
hippocampal neurons18. Since immature granule neurons play a 17. Nakano, M., Imamura, H., Nagai, T. & Noji, H. Ca2+ regulation of
role in hippocampal-dependent memory48, their reduction may mitochondrial ATP synthesis visualized at the single cell level. ACS Chem.
contribute to the memory impairment in LV-G1-1N3R-injected Biol. 6, 709–715 (2011).
mice. PV interneurons also play a key role in hippocampal func- 18. Sultan, S. et al. Synaptic integration of adult-born hippocampal neurons is
locally controlled by astrocytes. Neuron 88, 957–972 (2015).
tion. By exerting an important control over granule neurons, they
19. Crosby, K. C. et al. Nanoscale subsynaptic domains underlie the organization
fine-tune their activity and enable pattern separation. Furthermore, of the inhibitory synapse. Cell Rep. 26, 3284–3297 (2019).
PV interneurons enable the generation of gamma oscillations, 20. Cardin, J. A. et al. Driving fast-spiking cells induces gamma rhythm and
which support coincidence detection and regulate circuit perfor- controls sensory responses. Nature 459, 663–667 (2009).
mance49. The strong reduction of PV immunostaining in LV-G1- 21. Pöschel, B., Heinemann, U. & Draguhn, A. High-frequency oscillations in the
dentate gyrus of rat hippocampal slices induced by tetanic stimulation. Brain
1N3R-injected mice suggests that PV interneurons likely underlie Res. 959, 320–327 (2003).
their memory impairments, a possibility that is further supported 22. Towers, S. K. et al. Fast network oscillations in the rat dentate gyrus in vitro.
by the NRG1p-mediated behavioral rescue. J. Neurophysiol. 87, 1165–1168 (2002).
Astrocytes are key actors in brain physiology and play a fun- 23. Espinoza, C., Guzman, S. J., Zhang, X. & Jonas, P. Parvalbumin+ interneurons
damental role in the regulation of neural functioning50 and adult obey unique connectivity rules and establish a powerful lateral-inhibition
microcircuit in dentate gyrus. Nat. Commun. 9, 4605 (2018).
neurogenesis18. Our observations suggest that these cells may play 24. Gillespie, A. K. et al. Apolipoprotein E4 causes age-dependent disruption of
a greater role than expected in AD. Although the extent to which slow gamma oscillations during hippocampal sharp-wave ripples. Neuron 90,
astrocytes are involved in the etiology of AD remains unclear, our 740–751 (2016).

Nature NeuroscIence | www.nature.com/natureneuroscience


NATuRE NEuRoScIEncE Articles
25. Hu, H., Gan, J. & Jonas, P. Fast-spiking, parvalbumin+ GABAergic 38. Dixit, R., Ross, J. L., Goldman, Y. E. & Holzbaur, E. L. F. Differential
interneurons: from cellular design to microcircuit function. Science 345, regulation of dynein and Kinesin motor proteins by tau. Science 319,
1255263 (2014). 1086–1089 (2010).
26. Marissal, T. et al. Restoring wild-type-like CA1 network dynamics and 39. Amadoro, G. et al. AD-linked, toxic NH2 human tau affects the quality
behavior during adulthood in a mouse model of schizophrenia. Nat. Neurosci. control of mitochondria in neurons. Neurobiol. Dis. 62, 489–507 (2014).
21, 1412–1420 (2018). 40. Yoshiyama, Y., Zhang, B., Bruce, J., Trojanowski, J. Q. & Lee, V. M.-Y. Reduction
27. LoPresti, P., Szuchet, S., Papasozomenos, S. C., Zinkowski, R. P. & Binder, L. of detyrosinated microtubules and Golgi fragmentation are linked to
I. Functional implications for the microtubule-associated protein tau: tau-induced degeneration in astrocytes. J. Neurosci. 23, 10662–10671 (2003).
localization in oligodendrocytes. Proc. Natl Acad. Sci. USA 92, 10369–10373 41. van Bergeijk, P., Adrian, M., Hoogenraad, C. C. & Kapitein, L. C. Optogenetic
(1995). control of organelle transport and positioning. Nature 518, 111–114 (2015).
28. Müller, R., Heinrich, M., Heck, S., Blohm, D. & Richter-Landsberg, C. 42. Forman, M. S. Transgenic mouse model of tau pathology in astrocytes
Expression of microtubule-asssciated proteins MAP2 and tau in cultured rat leading to nervous system degeneration. J. Neurosci. 25, 3539–3550 (2005).
brain oligodendrocytes. Cell Tissue Res. 288, 239–249 (1997). 43. Piacentini, R. et al. Reduced gliotransmitter release from astrocytes mediates
29. Boisvert, M. M., Erikson, G. A., Shokhirev, M. N. & Allen, N. J. The aging tau-induced synaptic dysfunction in cultured hippocampal neurons. Glia 65,
astrocyte transcriptome from multiple regions of the mouse brain. Cell Rep. 1302–1316 (2017).
22, 269–285 (2018). 44. Whalley, K. Neurodegenerative disease: spreading the tau. Nat. Rev. Neurosci.
30. Yamada, K. et al. In vivo microdialysis reveals age-dependent decrease of 10, 548–548 (2009).
brain interstitial fluid tau levels in P301S human tau transgenic mice. J. 45. Hainmueller, T. & Bartos, M. Dentate gyrus circuits for encoding, retrieval
Neurosci. 31, 13110–13117 (2011). and discrimination of episodic memories. Nat. Rev. Neurosci. 21, 1–16 (2020).
31. Sanders, D. W. et al. Distinct tau prion strains propagate in cells and mice 46. Cope, E. C. & Gould, E. Adult neurogenesis, glia and the extracellular matrix.
and define different tauopathies. Neuron https://doi.org/10.1016/j. Cell Stem Cell 24, 690–705 (2019).
neuron.2014.04.047 (2014) 47. Moreno-Jiménez, E. P. et al. Adult hippocampal neurogenesis is abundant in
32. Perea, J. R. et al. Extracellular monomeric tau is internalized by astrocytes. neurologically healthy subjects and drops sharply in patients with Alzheimer’s
Front. Neurosci. 13, 442 (2019). disease. Nat. Med. 25, 554–560 (2019).
33. Dujardin, S. et al. Ectosomes: a new mechanism for non-exosomal secretion 48. Toda, T., Parylak, S. L., Linker, S. B. & Gage, F. H. The role of adult
of tau protein. PLoS ONE 9, 28–31 (2014). hippocampal neurogenesis in brain health and disease. Mol. Psychiatry
34. Goetzl, E. J. et al. Cargo proteins of plasma astrocyte-derived exosomes in https://doi.org/10.1038/s41380-018-0036-2 (2018)
Alzheimer’s disease. FASEB J. 30, 3853–3859 (2016). 49. Sohal, V. S., Zhang, F., Yizhar, O. & Deisseroth, K. Parvalbumin neurons and
35. Wang, Y. & Mandelkow, E. Tau in physiology and pathology. Nat. Rev. gamma rhythms enhance cortical circuit performance. Nature 459, 698–702
Neurosci. 17, 5–21 (2016). (2009).
36. Ferrer, I. et al. Aging-related tau astrogliopathy (ARTAG): not only tau 50. Bazargani, N. & Attwell, D. Astrocyte calcium signaling: the third wave. Nat.
phosphorylation in astrocytes. Brain Pathol. 28, 965–985 (2018). Neurosci. 19, 182–189 (2016).
37. Goode, B. L., Chau, M., Denis, P. E. & Feinstein, S. C. Structural and
functional differences between 3-repeat and 4-repeat tau isoforms: Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
implications for normal tau function and the onset of neurodegenerative published maps and institutional affiliations.
disease. J. Biol. Chem. 275, 38182–38189 (2000). © The Author(s), under exclusive licence to Springer Nature America, Inc. 2020

Nature NeuroscIence | www.nature.com/natureneuroscience


Articles NATuRE NEuRoScIEncE

Methods Primary rat hippocampal cultures. For co-cultures, timed-pregnant rats


Human participants. qaBrains were collected with individuals’ informed consent (RjHan:WI, Janvier) were killed by CO2 inhalation, and E17 embryos were
and the authorization of the Ethics Commission of the Lausanne University collected in Petri dishes containing Hank’s balance salt solution (HBSS; Gibco, Life
Hospital and CHU of Lille. A total of 30 brains were examined: 21 from individuals Technologies) to enable hippocampal dissection. Cells were prepared following
with sporadic AD without known familial history and 9 from control individuals published protocols66. The cells were plated at a density of 3 × 105 cells per cm2
who showed no sign of neurological disorder and who were age- and sex-matched in neuronal medium in multiwell dishes. For astrocyte cultures, P1 rat pups
to individuals with AD (Supplementary Table 1 and Extended Data Fig. 1a–c). All were used (Charles River), and cells were plated at a density of 20,000 cells per
individuals with AD had been hospitalized in the Service of Old Age Psychiatry cm2 in multiwell dishes. Hippocampal co-cultures and primary hippocampal
of the Lausanne University hospital or CHU of Lille and diagnosed according astrocytes were infected at DIV4 and DIV8 respectively, with 0.6 pg p24
to the revised Diagnostic and Statistical Manual of Mental Disorders, Third antigen per cell of LV-G1-CFP, LV-G1-GFP, LV-G1-1N3R, LV-G1-MitoTimer
Edition (DSM-IIIR) criteria. Clinical diagnosis and Braak stage were confirmed or LV-G1-MitoGoAteam2, corresponding to 1.2 pg of p24 antigen for a double
by postmortem neuropathological examination in the Department of Pathology infection and 1.8 ng for a triple infection.
of the Lausanne University Hospital for each case, following previously described
protocols51–53. Tau phosphorylation was assessed using an antibody specific for Dot-blot procedure. Increasing concentrations (0 to 500 ng in 200 µl PBS) of
the AT8 epitope in the anterior hippocampus, in the prefrontal, parietal and recombinant tau (3R or 4R) were loaded onto nitrocellulose membrane (0.4 µm).
temporal associative isocortex and in the primary visual cortex. The brains were Recombinant tau proteins were produced as described previously67. The membrane
removed with a postmortem delay of no more than 60 h and stored in buffered 10% was incubated 1 h at room temperature (RT) in a blocking solution containing
formaldehyde until sampling. 5% milk-TNT (15 mM Tris, 140 mM NaCl and 0.05% Tween 20) and transferred
to the antibody solution diluted in 5% milk-TNT (mouse monoclonal antibody
Immunohistochemistry on human samples. Blocks of ~5 mm × 5 mm of the against 3R tau (RD3, Millipore; 1/2,000) or mouse monoclonal antibody against 4R
dorsal hippocampus were embedded in paraffin and cut into 20-µm-thick sections. tau (RD4, Millipore; 1/1,000)) for incubation overnight at 4 °C. Tau proteins were
To reduce autofluorescence, we followed a recently developed protocol47,54. Briefly, revealed by enhanced chemiluminescence (ECL; GE Healthcare) using horseradish
samples were incubated in a 0.5% sodium borohydride (NaBH4; Sigma-Aldrich, peroxidase (HRP)-conjugated anti-mouse secondary antibody (Vector, 1/50,000).
213462) solution, followed by a citrate buffer antigen retrieval (HC-AR) step. Then, A negative control without the primary antibodies was used to exclude nonspecific
sections were microwaved for 10 min in tris-buffered saline and then incubated signal.
for 30 min in 95% formic acid (Sigma Chemical). Slices were then rinsed and
incubated 1 h in normal serum (rabbit or swine). Next, slices were immersed for Protein extraction and immunoblotting. Mouse hippocampus and cortex were
48 h at 4 °C in the following primary antibodies: (mouse anti-AT8; Thermo Fisher, dissected and suspended in PBS to a final concentration of 1 µg µl−1. Next, 10 µg
MN1020; 1:500 dilution), (mouse anti-AD2; Bio-Rad, 56484; 1:250), (mouse of sample was loaded onto a 4–12% Bis-Tris (Criterion gel, Bio-Rad), followed by
anti-RD3; Merck, 05-803; 1:75), (mouse anti-RD4; Merck, 05-804; 1:150), (rabbit transfer onto a 0.45-µm nitrocellulose membrane. After three rinses, membranes
anti-S100β; ab41548; 1:500), (mouse anti-PSD95; ABR, MA1-045; 1:75), (mouse were incubated with a blocking solution for 30 min at RT before incubation with
anti-amyloid-β; DAKO, M0872; 1:100), (mouse anti-synaptophysin; ab8049; rabbit polyclonal anti-V5 (Millipore, AB3792; 1:10,000) overnight at 4 °C. The
1:250) for 24 h at 4 °C. Then, slices were rinsed and incubated for 24 h at 4 °C in membrane was then incubated with the secondary anti-rabbit HRP antibody for
the following secondary antibodies: Alexa Fluor 488-, 555- or 647-conjugated 45 min at RT (Vector; 1:5,000). Signal was visualized using ECL western blotting
highly cross-adsorbed donkey anti-goat, rabbit, mouse, rat or donkey anti-mouse detection reagents (GE Healthcare) in an Amersham Imager 600.
biotinylated. Either the primary or the secondary antibody was omitted in
negative-control sections. All sections were counterstained for 10 min with DAPI Animals and stereotaxic delivery of LV. Three-month-old male C57BL/6 mice
(Merck; 1:5,000 dilution) to label nuclei. Immunohistochemistry was followed by were purchased from Janvier. All animals were housed in a temperature-controlled
a final autofluorescence elimination step, using an autofluorescence eliminator room (22 °C ± 1 °C) and maintained on a normal 12-h light/dark cycle with access
reagent (EMD Millipore, 2160) and following the manufacturer’s instructions. to food and water ad libitum. Mice were separated into cages of four mice per
Samples were mounted in VECTASHIELD Antifade Mounting Medium with DAPI cage. All procedures were performed in accordance with the overly strict Swiss
(Vectors Lab, H-1200) and stored at 4 °C until analysis. legislation on the care and use of laboratory animals. Mice were anesthetized by
intraperitoneal injection of a mixture of 100 mg kg−1 ketamine (Ketasol, Graeub)
Quantification of human samples. Quantification of AD2+ cells, Aβ plaques, and 10 mg kg−1 xylazine (Rompun, Bayer Health Care). The animals received 2 μl
PSD95 and synaptophysin optical density was performed on a minimum of of LV bilaterally injected into the dorsal dentate gyrus at the coordinates: ±1.5 mm
four sections (separated by at least 160 µm) per individual, using a digital lateral to the midline, −2 mm posterior to bregma and −2.4 mm ventral to the dura
camera (3CCD Hitachi HV-F202SCL) mounted on a slide scanner microscope relative to bregma. The LV were injected at 0.2 μl min−1 and the needle was left in
(×20 objective; Zeiss axioscan Z1). AD2 inclusions were analyzed only when place for 5 min. Animals received injections of the analgesic buprenorphine at a
their surface was between 150 µm2 and 2,000 µm2 and Aβ plaques between 350 dose of 0.1 ml per 100 g after injection.
µm2 and 5000 µm2. To determine the presence of 3R and 4R tau inclusion in
s100β-expressing astrocytes, we analyzed 15–20 stacks/individuals with AD, 40–60 Immunohistochemistry on mouse samples. For injected animals, mice were
S100β+ astrocytes/individual and tau isoform, for a total of 1,840 astrocytes for the deeply anesthetized and transcardially perfused with 4% paraformaldehyde. A
presence of 3R tau and 1,537 astrocytes for the presence of 4R tau. series of 1-in-6, 30-µm-thick coronal sections were prepared and incubated in
The volume of S100β+, 3R and 4R tau inclusions was determined using PBS containing 0.3% Triton X-100 with the following primary antibodies: chicken
autoregressive algorithms of the Imaris surface plugin55. Individual astrocytes were anti-GFP (Biotrend, 55423; 1:5,000 dilution), rabbit anti-CFP (Acris, TA332666;
considered to contain 3R or 4R tau inclusions when the volume of these inclusions 1:250), rabbit anti-GFAP (DAKO, M0761; 1:500), mouse anti-NeuN (Chemicon,
represented at least 5% of the volume of the soma. MAB377; 1:1,000), mouse anti-V5-Tag (Invitrogen, 46-0705; 1:1,000), goat
anti-V5-Tag (Abcam, ab9137; 1:1,000), GluR2/3 (Millipore, 07-598; 1:1,000),
Lentiviral vectors. We used a new VSV-G pseudotyped LV to selectively express mouse anti-BrdU (CBL187; 1:1,000), rabbit anti-VGAT (Synaptic Systems, 131003;
transgenes in astrocytes. The gfaABC1D promoter was kindly provided by M. 1:1,000), mouse anti-gephyrin (Synaptic Systems, 131011; 1:500), rabbit anti-c-Fos
Brenner56,57 and ligated to the B(3) enhancer to generate the G1 promoter56, which (Merck, ABE457; 1:500) and mouse anti -MC-1 (courtesy of Beat Riederer
was then cloned into the SIN-cPPT-gateway-WPRE-miR124T transfer plasmids, laboratory68, 1:250). After several rinses with PBST, sections were incubated
which contains four copies of the neuron-specific miRNA-124 target sequence for 90 min at RT in a PBST solution containing the following mix of secondary
(miR124T; full homology58) to repress transgene expression in neurons59, the antibodies: Alexa Fluor 488-, 555- or 647-conjugated highly cross-adsorbed
WPRE and central polypurine tract (cPPT) to increase transgene expression60,61, donkey anti-goat, rabbit, mouse, rat or guinea-pig (1:500; Life Technologies). All
and a 400-nucleotide deletion in the 3′ long terminal repeat (self-inactivating sections were counterstained for 10 min with DAPI (Merck; 1:5,000 dilution) to
vector) to increase biosafety62. Human 1N3R and 1N4R tau isoforms with a label nuclei. Immunohistochemistry against VGAT/gephyrin was followed by a
V5 tag (14 amino acids; GKPIPNPLLGLDST inserted between the sequences final autofluorescence elimination step. To this end, autofluorescence eliminator
encoding exons two and four)33,63 and the CFP reporter gene (Takara Bio reagent (EMD Millipore, 2160) was used, per the manufacturer’s instructions.
Europe), were used to generate the SIN-cPPT-GfaABC1D(B)3-tau-1N3R-V5 Samples were mounted in VECTASHIELD Antifade Mounting Medium with DAPI
and SIN-cPPT-GfaABC1D(B)3-tau-1N4R-V5 (hereafter called LV-G1-1N3R (Vectors Lab, H-1200) and stored at 4 °C until analysis.
and LV-G1-1N4R), SIN-cPPT-GfaABC1D(B)3-GFP-WPRE-miR124T and
SIN-cPPT-GfaABC1D(B)3-CFP-WPRE-miR124T (hereafter called LV-G1-GFP Quantification of cell populations in the mouse hippocampus. Quantification
and LV-G1-CFP) plasmids. MitoTimer was kindly provided by A. Terskikh64. The of DAPI+, GFP+, PV+, GluR2/3+, NeuN+, V5+, DCX+ and BrdU+ cells per brain
mitochondrial ATP sensor was kindly provided by H. Noji17. LVs were produced was performed from 1-in-6 sections spaced 300 µm apart using a digital camera
as previously reported65, using the packaging plasmid pCMV∆ R8.92, the transfer (3CCD Hitachi HV-F202SCL) on a slide scanner microscope (×20 objective, Zeiss
vector expressing the transgenes, pRSV-Rev and pMD.2G (VSV-G envelope). The axioscan Z1). In hippocampal section containing infected cells, immunopositive
final viral concentration was calculated by the p24 enzyme-linked immunosorbent cells located in the region of interest (ML, GCL, subgranular layer, CA1, CA3,
assay (RETROtek). cortex and hilus) were counted using image analysis software Zen 2 (black 8.0

Nature NeuroscIence | www.nature.com/natureneuroscience


NATuRE NEuRoScIEncE Articles
edition and blue 2012 edition). Cell density was calculated by dividing the total donkey anti-mouse (Molecular Probes, A31571) were applied (at a concentration
number of cells for each acquisition by the surface of each area of interest. For of 1:500) for 30 min at RT. Coverslips were then imaged using a Leica SP5 confocal
PV labeling intensities, 100–150 cells were quantified in arbitrary units as the microscope.
mean of all isolated pixels of soma. Each optical density was normalized via the
subtraction of a slide section in which signal was absent (black). Normalization and MitoTimer mitochondrial system analyses in vitro. The astrocytic mitochondrial
recalibration across different experiments was achieved by using internal control system was assessed 10 d after viral infection. Cells were imaged by acquisition of
animals. Animals for the quantification of c-Fos+ cells were perfused 90 min after multiple optical sections with a Zeiss LSM 710 Quasar confocal system.
memory test (object location task).
Morphology and localization. Confocal images were imported into Imaris XT
Quantification and determination of cell phenotype in vitro and in vivo. Cell (Bitplane AG). Green (500–540 nm) and red (580–640 nm) channels from
phenotypes were determined from 350 GFP+ cells per mouse or 50 per neuron– MitoTimer were merged using the Imaris channel arithmetic MATLAB plugin
glial culture. Colocalization with GFAP, S100b, NeuN or IBA1 was assessed by to visualize whole mitochondria in CFP+ cells and mitochondrial volume
confocal microscopy (×40 oil immersion objective, Zeiss LSM 710 Quasar) over reconstruction was performed using the Imaris surface plugin55. Using the CFP
the entire z axis. Labeled cells were rotated in orthogonal planes (x and y) to channel to visualize the entire cell, a 20-µm diameter circle was drawn around
verify double labeling. All analyses were performed in sequential scanning mode the center of the soma of astrocytes. Mitochondria found within this circle were
to prevent crossover between channels. The estimated fraction of GFP+ cells considered as proximal and mitochondria found further away were considered as
co-labeled with NeuN or GFAP was calculated for each animal. Absolute numbers distal. Mitochondrial distribution and morphology (length, width and surface)
of GFP+GFAP+, GFP+S100b+ and GFP+NeuN+ cells were obtained by multiplying were automatically counted for each compartment and the frequency distribution
the corresponding estimated fraction of co-labeled GFP+ cells by the total number calculated in ‘proximal’ and ‘distal’ processes for each cell.
of GFP+ cells for each animal. GFP+ cells located in dentate gyrus were classified as
astrocytes, RGL and neurons based on morphology. Astrocytes were characterized Motility. For mitochondrial motility, single mitochondria were manually followed
by a large spheroid or pyramidal soma with ramified processes. Neurons displayed for each time point from live imaging acquisition based on merged green and
an oval-shaped soma with an apical dendritic tree extending through the GCL red channels. The mean velocity and total track length were then automatically
and reaching the ML. RGL cells displayed a prototypical morphology, including calculated by Imaris. The total track length traveled (Δ) was used to classify
a nucleus located in the subgranular zone of the dentate gyrus, a radial process mitochondria as highly mobile (Δ ≥ 20 µm), mobile (6 µm ≤ Δ ≤ 20 µm) or
extending through the GCL and extensively branching into the outer GCL and the stationary (6 µm ≤ Δ). The direction of each mitochondria was determined by
ML, and a few basal processes extending towards the hilus69,70. visualizing the displacement vector and was defined as anterograde when the
mitochondria moved towards the periphery of the astrocyte and retrograde when it
Morphological analyses of GFP+ astrocytes in vitro and in vivo. Approximately moved towards the soma.
15–20 hilar GFP+ astrocytes per mouse (or 50 GFP+ astrocytes in vitro) were
imaged with a Zeiss LSM 880 Quasar confocal system (63× + 2× numerical zoom) MitoTimer redox state ratio. The red and green mean intensities were automatically
equipped with Airyscan. Care was taken to only image astrocytes with a soma calculated in the soma and proximal and distal processes based on green and red
entirely contained within the thickness of the section. Images consisted of 50–75 merged channel volume reconstruction of MitoTimer using the Imaris statistical
optical sections (z = 0.3 µm). Three-dimensional reconstructions of a series of plugin. The fluorescence ratio (R555/488) was normalized against of the fluorescence
confocal images were deconvolved (Huygens SVI) and analyzed using Imaris XT the control condition (LV-G1-CFP) for each culture.
(Bitplane AG) and the ‘autopath’ algorithm of the ‘filament’ plugin. Soma volume
was calculated using the Imaris surface plugin55 and was manually corrected to FRET mitochondrial ATP imaging. The ATP mitochondrial system was assessed
exclude the main processes. The NIVs were calculated as previously described71. 10 d after viral infection. Astrocytes were recorded every 5 s for 5 min using a Zeiss
Briefly, for every astrocyte analyzed (20–25 cells per group), three randomly LSM 710 Quasar confocal microscope with a ×100 objective and digital zoom set
chosen regions of interest of 15 µm × 15 µm × 10 µm, devoid of soma and large to 4. The excitation wavelength was 350 nm for CFP and 488 nm for GFP, with
branches were imaged. Astrocytic processes were 3D reconstructed in the hilus detection of blue (410–480 nm), GFP (493–545 nm) and OFP (580–640 nm). The
using Imaris software and their volume was measured. OFP/GFP ratio was calculated by dividing the OFP mean intensity by the GFP
intensity for ten regions of interest in the soma and proximal and distal CFP+
Quantification of gephyrin and VGAT punctae. Images were acquired in the processes of astrocytes.
vicinity of distal processes of GFP+ astrocytes that were entirely contained within
the section thickness. Typically, 40–50 images per group (around 10 per animals) Calcium imaging. The intracellular calcium concentration was assessed 11 d
were acquired (series of 50–75 multiple optical sections, z = 0.2 µm) with a Zeiss after viral infection. Cultures were loaded with 5 µM Fluo-4 AM (Invitrogen) for
LSM 880 Quasar confocal system (63× + 4× numerical zoom) equipped with 15 min in the presence of 0.02% Pluronic F-127 (Invitrogen) at 37 °C and 5% CO2
Airyscan. Images were imported into Imaris XT (Bitplane AG) and corrected in the dark in a HEPES-KRH buffer at pH 7.4 and de-esterified for 10–15 min
for background. VGAT or gephyrin dots were determined using autoregressive before imaging. The mean fluorescence for Fluo-4 AM was normalized over CFP
algorithms of Spot plugin. The density of VGAT and gephyrin dots was calculated fluorescence and calculated for ten regions of interest in the soma and proximal
by dividing the total number of dots for each acquisition by the volume of interest. and distal CFP+ processes.
For the VGAT/gephyrin pairing analysis, we used colocalized Spot MATLAB script
with 0.3 µm for the closest distance between spots. The VGAT/gephyrin ratio was In vitro electrophysiology. Slice preparation. Mice (7 months old) were injected
then calculated. with viruses (LV-G1-1N3R, n = 6) or CFP (LV-G1-GFP, as controls, n = 6)
in the dentate gyrus and anesthetized with ketamine/xylazine, and perfused
MitoTimer mitochondrial analyses in the mouse hippocampus. The with cold oxygenated sucrose-containing artificial cerebrospinal fluid (ACSF;
hippocampal astrocytic mitochondrial system was assessed four months after 252 mM sucrose, 3 mM KCl, 2 mM MgSO4, 1.2 mM CaCl2, 1.2 mM NaH2PO4,
co-infection with LV-G1-CFP + LV-G1-MitoTimer or LV-G1-CFP + LV-G1- 24 mM NaHCO3 and 10 mM glucose; pH 7.4) for 10 min before decapitation.
1N3R + LV-G1-MitoTimer. Multiple optical sections (z = 0.3 µm) of confocal Three successive coronal brain slices (400-µm thick) containing both dorsal
images were acquired throughout the section of cells located in the polymorphic hippocampi (bregma: −1.5 mm to −2.7 mm) were prepared with a vibroslicer in
and subgranular layers of the dentate gyrus, with a Zeiss LSM 780 Quasar confocal cold sucrose-containing ACSF and placed in oxygenated recovery ACSF at 36 °C
system (63× + 4× numerical zoom). Fluorescence images were captured using for ~15 min. This recovery solution was a N-methyl-d-glucamine (NMDG)/
similar mirror/filter, excitation and detection parameters as those used for the HEPES-based ACSF (93 mM NMDG, 20 mM HEPES, 2.5 mM KCl, 10 mM
in vitro experiments. For localizing processes within mitochondria, acquisitions MgSO4, 0.5 mM CaCl2, 1.2 mM NaH2PO4, 30 mM NaHCO3, 3 mM sodium
were imported into Imaris XT (Bitplane AG). The green (500–540 nm) and red pyruvate, 2 mM thiourea, 5 mM ascorbic acid and 25 mM glucose; pH 7.3). Slices
(580–640 nm) channels from MitoTimer were merged using the Imaris channel were then transferred into a Haas-type interface chamber and superfused with
arithmetic MATLAB plugin to visualize whole mitochondria. Mitochondrial recording ACSF (129 mM NaCl, 3 mM KCl,1.8 mM MgCl2, 1.6 mM CaCl2, 1.25 mM
volume reconstructions were performed using the Imaris surface plugin. To assess NaH2PO4, 21 mM NaHCO3 and 10 mM glucose; pH 7.4). After 1 h, the ACSF
the redox state of the mitochondria, the mean intensities of the red and green temperature was slowly raised from RT to 32 °C.
channels were automatically calculated using the Imaris statistics plugin and
normalized against those of the control condition (LV-G1-CFP). Recordings. Field potentials were recorded with ACSF-filled glass electrodes
(~5 MOhm). Signals were amplified (differential amplification mode, gain 1000×,
Immunohistochemistry on primary cultures. Primary cultures on glass coverslips NPI Ext-2 amplifiers), bandpass-filtered at 1–1,000 Hz and digitized at 2 KHz and
were fixed and immunostained with the following primary antibodies: chicken acquired with Spike2 software (CED). In each hippocampal slice (six per animal),
anti-GFP (Chemicon, AB16901; 1:500), rabbit anti-GFAP (Dako, Z0334; 1:1,000), electrodes were positioned at four different sites in the GCL–ML interface of the
rabbit anti-NeuN (Chemicon, MAB377; 1:1,000), mouse anti-V5 (Millipore, dentate gyrus (Fig. 6n). Transient neuronal activity and high-frequency oscillations
AB3792; 1:500), rabbit anti-CFP (Acris, TA332666; 1:250). Following washing, were evoked in the dentate gyrus by a short pressure injection (200 ms, ~7–12
Alexa Fluor 594 goat anti-mouse (Invitrogen, A11005) and Alexa Fluor 647 bars) of glutamate (10 mM in ACF) via a glass pipette (tip diameter: ~8–12 µm)

Nature NeuroscIence | www.nature.com/natureneuroscience


Articles NATuRE NEuRoScIEncE
placed into the hilus21. These experiments were performed blindly without The mouse was allowed to explore for 10 min, and the time spent exploring each
knowledge of the type of LV injected. After recording, slices were fixed with 4% object was recorded. The nature and position (left or right) of the new object was
paraformaldehyde for later verification by a second experimenter of the presence randomized. The open field was cleaned thoroughly between the introductions
of GFP-labeled astrocytes in the dentate gyrus of the recorded slices. Only slices of each mouse to eliminate olfactory cues. Memory for the familiar object was
displaying proper LV injection and expression in the dentate gyrus were used evaluated by calculating the preference index for the new object, expressed as the
for further analyses (for LV-G1-1N3R: 5 mice and 22 hippocampal slices; for percentage of time spent exploring the new object per total time spent exploring
LV-G1-GFP: 5 mice and 19 hippocampal slices). both objects. Independent groups of mice were used for the object location and
object recognition tasks, so that each animal was submitted to either one or the
Analysis. Time–frequency decompositions were performed using the MATLAB other task.
toolbox EEGLAB (version 14.1.1) on recording segments comprising the 2 s
before and the first 12 s following glutamate stimulation. Time–frequency Morris water maze. During the training phase, mice were placed in a pool filled
decompositions (from 10 Hz to 160 Hz, 1 Hz steps) were computed using Morlet with opaque water set at 25 °C. Training consisted of 6 d of four trials per day. The
waveform transforms (f0/σf of (3 0.5), 3-cycle wavelet with a slow linear increase platform was always hidden in the southeast quadrant of the pool, but the mice
(coefficient 0.5)). Data were assessed for the normality using the Kolmogorov– were released at various points around the swimming pool, with the point of release
Smirnov and Shapiro–Wilk tests, and for the homogeneity of variance using the being counterbalanced every day. The intertrial interval was between 10 and 20 min.
Levene test. When the data did not meet the criteria of normality, they were first Swimming tracks were recorded using video hardware and EthoVision software
log transformed before the statistical analyses. For each response type, the total (Noldus). Spatial memory was then assessed during a probe trial 24 h after the
power and the frequency at which the power was maximal were compared between final training (day 7). After establishing robust spatial preference for the platform
groups. Comparisons between groups were performed using univariate ANOVAs. location, either reversal trials were performed during which the platform was placed
The Welch t-test was used, however, when the homogeneity of variance was not in a different location. At day 8, mice started a new, 3-d training session (two trials
met. The initial voltage deflection following local glutamate application and per day), where the platform was located in a new position (northwest) to start
reflecting the overall induced activity was used as covariate. Statistical analysis was reversal learning. Swim paths were recorded and analyzed by a tracking system
performed with SPSS, and significance was set to P = 0.05. (EthoVision, Noldus). The assessed variables were escape latencies and, for the
probe trials, time spent in target quadrants and platform place proximity indices.
Behavioral procedures. Four months after viral injection, tests were performed During the probe tests, time spent in each quadrant and the numbers of crossings
in the following order: group of animals were tested for light-dark box (day 121), over the location of the platform (virtual circle) compared to the mean crossing
Y-maze (day 123), open field (day 127) and object recognition or location (day number of the three other virtual circles in the other three quadrants was calculated.
128) or for Morris water maze (days 120–128) and contextual fear conditioning
(days 130–131). For NRG1p injections, another group of mice received one Contextual fear conditioning. Mice were assessed for fear memory accuracy in
intraperitoneal injection of NRG1p (0.1 μg kg−1 in 0.9% saline; Prospec) or saline a fear-conditioning paradigm. For training, each mouse was introduced in a
solution 1 h before the object location test and were euthanized 90–100 min after conditioning chamber (FCS-NG 46000, Ugo Basile) measuring 19 × 10 × 30 cm,
the test. with a metal wire floor and transparent plastic wall, set in a white soundproof
cubicle (context A). The floor of the chamber under the grid was lined with tissue
Dark/light box. The dark/light box consisted of two compartments made with paper, which was changed between mice. After 3 min, mice received a single,
acrylic transparent glass, placed in the open-field arena, a black/dark compartment 2-s foot shock (0.5 mA) and were removed from the chamber 15 s later. The
(40 cm × 20 cm × 15 cm; 2 lux) and a white/illuminated compartment (same conditioned freezing response to context A was assessed 24 h later, upon 3-min
dimensions, 350 lux). Both compartments were connected by an aperture. Each exposure to exactly the same chamber. The freezing time, defined as the absence of
mouse was released in the same corner of the illuminated compartment, and the all movements with the exception of those related to respiration, was recorded by
number of exits and total time in the lit compartment were recorded for 6 min. overhead cameras and measured using automated scoring systems (AnyMaze).

Y-maze test. This test was performed as previously described72. The symmetrical Sample sizes, calculations and statistical analysis. Sample sizes are indicated in
Y-maze, made of acrylic glass, consisted of three arms, each 40-cm long, 15-cm the legend of the corresponding figures. Human sample size was not predicted. We
high and 5-cm wide. Each mouse was placed in the center of the Y-maze and was have used a collection of human samples composed of 9 healthy control individuals
free to explore the arena for 6 min. After each session, the maze was thoroughly and 21 individuals with AD. For cellular and behavioral assays, the sample size was
cleaned using ethanol and water and dried. The number of entries was recorded chosen to account for statistical variability of cultures (more than three cultures)
for each mouse while observing the mouse via a camera; one entry was defined as and surgical and behavioral procedures (more than eight animals), based on
both hind paws of the animal being completely inside the arm. The measure for previous studies69,77. Human samples were classified on the basis of neurological
working memory was the percentage of alternations, that is, the number of triads and neuropathological examination, in particular on the presence of tau and
divided by the maximum possible alternations (the total number of entries minus Aβ in the hilus. The order of culture and mouse used for infection, injection
2) × 100 (ref 73). and behavioral procedures was randomized for each experiment. Investigators
were blinded to group allocation when processing the tissue, performing cell
Object location test. This task is based on the spontaneous tendency of rodents counts and during confocal image acquisition and behavioral tests. The only
previously exposed to two identical objects to preferentially explore the object reasons for exclusion were problems encountered during culture (such as culture
that has been placed in a new location, rather than the non-displaced object74. The contamination) or failure of the injection procedure (no fluorescence observed in
day before the exploration phase, each mouse was placed in an open-field arena the hippocampus). Values are presented as the mean ± s.e.m.; N corresponds to
(35 cm × 34 cm × 40-cm high wall with a spatial pattern inside) for habituation the number of independent experiments and n to the overall number of values.
and allowed to explore the arena for 10 min. The total distance traveled in the Statistical analyses were performed on raw data with GraphPad Prism software
open field was measured by video tracking (Noldus EthoVision), to assess general v8.0. The normality of the data was verified using a Shapiro test. Data containing
motricity and activity. The next day, two identical objects were placed in the two experimental groups were analyzed using Student’s t-test (parametric
middle of the open-field arena, and mice were allowed to explore them for 10 min. observations), Mann–Whitney test (non-parametric observations), one-way and
The time exploring the two objects was scored. Spatial memory was tested 24 h two-way ANOVA tests and Wilcoxon matched pairs test (non-parametric paired
later when one of the objects (left or right counterbalanced) was moved to a observations), followed by Tukey’s post hoc analyses. Statistical analyses on data
new position. Mice were allowed to explore for 10 min. The time exploring the containing more than two experimental groups were performed using two-way
displaced object was calculated as the percentage of the total time exploring both ANOVA test, followed by Dunnett’s post hoc analyses, to account for multiple
objects. comparisons.
Object recognition test. This task is based on the spontaneous preference of rodents Reporting Summary. Further information on research design is available in the
for novelty and their ability to remember previously encountered objects75,76. The Nature Research Reporting Summary linked to this article.
procedure, equipment and analyses were similar to those described for the new
object location test, but the pattern inside the arena was removed. One day after
habituation, two identical objects were placed in the middle of the open field, and Data availability
the time the animal spent exploring each object was recorded. We ensured that The data that support the findings of this study are available from the
every mouse spent the same amount of time exploring the objects and avoided any corresponding author upon request. The map sequence for LV construction and
bias due to differences in individual levels of exploration by removing the animal microscopy acquisition data have been deposited in Zenodo.org at https://doi.
once it had explored the objects for a cumulative total of 30 s. Animals that did org/10.5281/zenodo.3953694. Source data are provided with this paper.
not achieve this criterion within 10 min were excluded (two animals). Recognition
memory was tested 24 h after the exploration phase. Mice were reintroduced into
the arena and exposed to two objects, a familiar object and a new object, for which References
the positions of the two objects were identical to those of session 1. The familiar 51. Braak, H. & Braak, E. Morphology of Alzheimer disease. Fortschr. Med. 108,
object was a triplicate copy of the sample used in session 1, to avoid olfactory trails. 624–624 (1990).

Nature NeuroscIence | www.nature.com/natureneuroscience


NATuRE NEuRoScIEncE Articles
52. Braak, H. & Braak, E. Staging of Alzheimer’s disease-related neurofibrillary 73. Wall, P. & Messier, C. Infralimbic kappa opioid and muscarinic M1 receptor
changes. Neurobiol. Aging 16, 271–278 (1995). interactions in the concurrent modulation of anxiety and memory.
53. Braak, H., Alafuzoff, I., Arzberger, T., Kretzschmar, H. & Tredici, K. Staging Psychopharmacol. 160, 233–244 (2002).
of Alzheimer disease-associated neurofibrillary pathology using paraffin 74. Ennaceur, A., Neave, N. & Aggleton, J. P. Spontaneous object recognition and
sections and immunocytochemistry. Acta Neuropathol. https://doi. object location memory in rats: the effects of lesions in the cingulate cortices,
org/10.1007/s00401-006-0127-z (2006). the medial prefrontal cortex, the cingulum bundle and the fornix. Exp. Brain
54. Flor-García, M. et al. Unraveling human adult hippocampal neurogenesis. Res. 113, 509–519 (1997).
Nat. Protoc. 15, 668–693 (2020). 75. Ennaceur, A. & Delacour, J. A new one-trial test for neurobiological studies
55. Richetin, K. et al. Amplifying mitochondrial function rescues adult of memory in rats. 1: behavioral data. Behav. Brain Res. 31, 47–59 (1988).
neurogenesis in a mouse model of Alzheimer’s disease. Neurobiol. Dis. 102, 76. Dodart, J. C., Mathis, C. & Ungerer, A. Scopolamine-induced deficits in a
113–124 (2017). two-trial object recognition task in mice. Neuroreport 8, 1173–1178 (1997).
56. Lee, Y., Messing, A., Su, M. & Brenner, M. GFAP promoter elements required 77. Richetin, K. et al. Genetic manipulation of adult-born hippocampal neurons
for region-specific and astrocyte-specific expression. Glia 56, 481–493 (2008). rescues memory in a mouse model of Alzheimer’s disease. Brain 138,
57. De Leeuw, B. et al. Increased glia-specific transgene expression with glial 440–455 (2015).
fibrillary acidic protein promoters containing multiple enhancer elements. J.
Neurosci. Res. 83, 744–753 (2006). Acknowledgements
58. Merienne, N. et al. Gene transfer engineering for astrocyte-specific silencing This study was supported by a Synapsis Foundation fellowship awarded to K.R.
in the CNS. Gene Ther. https://doi.org/10.1038/gt.2015.54 (2015) and the Lausanne University Hospital (CHUV) and by the Swiss National Science
59. Colin, A. et al. Engineered lentiviral vector targeting astrocytes in vivo. Glia Foundation (31003A_173128 to N.T. and K.R.). L.B., M.C., S.H., R.C. and S.E. were
57, 667–679 (2009). supported by the Programme Investissement d’avenir LabEx (laboratory excellence),
60. Déglon, N. et al. Self-inactivating lentiviral vectors with enhanced transgene DISTALZ (Development of Innovative Strategies for a Transdisciplinary approach to
expression as potential gene transfer system in Parkinson’s disease. Hum. ALZheimer’s disease), France Association PSP, the LiCEND (Lille Centre of Excellence
Gene Ther. 11, 179–190 (2000). in Neurodegenerative Disorders), CNRS, Inserm, Métropole Européenne de Lille,
61. Sirven, A. et al. The human immunodeficiency virus type-1 central DNA flap Univ. Lille, FEDER and DN2M. The authors thank the Cellular Imaging Facility of the
is a crucial determinant for lentiviral vector nuclear import and gene University of Lausanne for their technical support; F. Magara at the Center for Behavioral
transduction of human hematopoietic stem cells. Blood 96, 4103–4110 (2000). Studies of the Lausanne University Hospital, for assistance with the behavioral testing;
62. Zufferey, R. et al. Self-inactivating lentivirus vector for safe and efficient and H. Imamura of Kyoto University for the MitoGoAteam2 plasmid. We warmly
in vivo gene delivery. J. Virol. 72, 9873–9880 (1998). thank C. Rampon and M.C. Miquel at the University of Toulouse and G. Vachey, M.
63. Lobbestael, E. et al. Immunohistochemical detection of transgene expression Humbert-Claude, L. Tenenbaum and R. Jenni at the Lausanne University Hospital for
in the brain using small epitope tags. BMC Biotechnol. 10, 16 (2010). their precious help. We also thank S. Sultan, F. Cassé and T. Larrieu for their critical
64. Terskikh, A. et al. ‘Fluorescent timer’: protein that changes color with time. reading of the manuscript and helpful comments.
Science 290, 1585–1588 (2000).
65. Hottinger, A. F., Azzouz, M., Déglon, N., Aebischer, P. & Zurn, A. D.
Complete and long-term rescue of lesioned adult motoneurons by Author contributions
lentiviral-mediated expression of glial cell line-derived neurotrophic factor in K.R. conceived the project and co-supervised the study, acquired and analyzed
the facial nucleus. J. Neurosci. 20, 5587–5593 (2000). the data and wrote the manuscript. G.L. collected human samples and performed
66. Kaech, S. & Banker, G. Culturing hippocampal neurons. Nat. Protoc. 1, immunostainings. M.P. and R.P. acquired and analyzed microscopy data. M.M. acquired
2406–2415 (2006). the data for LV tropism. P.B. designed the calcium imaging experiments. P.S. and
67. Qi, H. et al. Nuclear magnetic resonance spectroscopy characterization of K.D. designed and performed the electrophysiology experiments. M.R. cloned the
interaction of tau with DNA and its regulation by phosphorylation. plasmids and produced the LV. C.P., E.P. and R.C. produced the in vitro cultures and
Biochemistry 54, 1525–1533 (2015). immunohistochemistry. S.H., S.B. and M.C. acquired and analyzed data. M.C. and L.B.
68. Jicha, G. A., Bowser, R., Kazam, I. G. & Davies, P. Alz-50 and MC-1, a new helped with the research design and critically revised the manuscript. N.T. designed and
monoclonal antibody raised to paired helical filaments, recognize supervised the study and wrote the manuscript. N.D. designed the LVs and supervised
conformational epitopes on recombinant tau. J. Neurosci. Res. 48, 128–132 the study.
(1997).
69. Richetin, K., Petsophonsakul, P., Roybon, L., Guiard, B. P. B. P. & Rampon, C. Competing interests
Differential alteration of hippocampal function and plasticity in females and The authors declare no competing interests.
males of the APPxPS1 mouse model of Alzheimer’s disease. Neurobiol. Aging
57, 220–231 (2017).
70. Gebara, E. et al. Heterogeneity of radial glia-like cells in the adult Additional information
hippocampus. Stem Cells 34, 997–1010 (2016). Extended data is available for this paper at https://doi.org/10.1038/s41593-020-00728-x.
71. Stogsdill, J. A. et al. Astrocytic neuroligins control astrocyte morphogenesis Supplementary information is available for this paper at https://doi.org/10.1038/
and synaptogenesis. Nature 551, 192–197 (2017). s41593-020-00728-x.
72. Holcomb, L. A. et al. Behavioral changes in transgenic mice expressing both
amyloid precursor protein and presenilin-1 mutations: lack of association Correspondence and requests for materials should be addressed to K.R. or N.T.
with amyloid deposits. Behav. Genet. 29, 177–185 (1999). Reprints and permissions information is available at www.nature.com/reprints.

Nature NeuroscIence | www.nature.com/natureneuroscience

You might also like