Download as pdf or txt
Download as pdf or txt
You are on page 1of 34

Neuron

Article

Loss of mTOR-Dependent Macroautophagy


Causes Autistic-like Synaptic Pruning Deficits
Guomei Tang,1 Kathryn Gudsnuk,2 Sheng-Han Kuo,1 Marisa L. Cotrina,3,7 Gorazd Rosoklija,4,8 Alexander Sosunov,3
Mark S. Sonders,1 Ellen Kanter,1 Candace Castagna,1 Ai Yamamoto,1 Zhenyu Yue,6 Ottavio Arancio,3
Bradley S. Peterson,4,8 Frances Champagne,2 Andrew J. Dwork,3,4,8 James Goldman,3 and David Sulzer1,4,5,8,*
1Department of Neurology
2Department of Psychology
3Department of Pathology and Cell Biology
4Department of Psychiatry
5Department of Pharmacology

Columbia University Medical Center, New York, NY10032, USA


6Departments of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
7Center for Translational Neuromedicine, University of Rochester, Rochester, NY 14642, USA
8New York State Psychiatric Institute, New York, NY 10032, USA

*Correspondence: ds43@columbia.edu
http://dx.doi.org/10.1016/j.neuron.2014.07.040

SUMMARY ASD model mice (Zoghbi and Bear, 2012). It remains however
unclear why spine pathology occurs and how it is associated
Developmental alterations of excitatory synapses with the onset and progression of ASD-related symptoms.
are implicated in autism spectrum disorders (ASDs). Postnatal synaptic development in mammalian cerebral cor-
Here, we report increased dendritic spine density tex is a dynamic process involving concurrent formation and
with reduced developmental spine pruning in layer V elimination/pruning (Purves and Lichtman, 1980; Rakic et al.,
pyramidal neurons in postmortem ASD temporal 1986). Synapse formation exceeds pruning at early ages,
yielding excessive excitatory synapses essential for the assem-
lobe. These spine deficits correlate with hyperacti-
bly of neural circuits. Synaptic elimination subsequently outpa-
vated mTOR and impaired autophagy. In Tsc2+/
ces formation, resulting in net spine pruning from childhood
ASD mice where mTOR is constitutively overactive, through adolescence. Consistently, the density of dendritic
we observed postnatal spine pruning defects, spines peaks in early childhood and is followed by a steep
blockade of autophagy, and ASD-like social behav- decline during late childhood and adolescence to adult levels
iors. The mTOR inhibitor rapamycin corrected ASD- (Penzes et al., 2011), a process that provides selection and
like behaviors and spine pruning defects in Tsc2+/ maturation of synapses and neural circuits.
mice, but not in Atg7CKO neuronal autophagy-de- While ASDs exhibit striking genetic and clinical heterogeneity,
ficient mice or Tsc2+/ :Atg7CKO double mutants. multiple ASD syndromes are caused by mutations in genes that
Neuronal autophagy furthermore enabled spine act to inhibit mammalian target of rapamycin (mTOR) kinase,
elimination with no effects on spine formation. Our including Tsc1/Tsc2, NF1, and Pten (Bourgeron, 2009). Synap-
tic mTOR integrates signaling from various ASD synaptic and
findings suggest that mTOR-regulated autophagy is
regulatory proteins, including SHANK3, FMRP, and the gluta-
required for developmental spine pruning, and activa-
mate receptors mGluR1/5 (Peça and Feng, 2012; Bourgeron,
tion of neuronal autophagy corrects synaptic pathol- 2009). Overactive mTOR signaling may produce an excess
ogy and social behavior deficits in ASD models with of synaptic protein synthesis, which could indicate a common
hyperactivated mTOR. mechanism underlying ASD. Synapses, however, must balance
protein synthesis and degradation to maintain homeostasis and
support plasticity (Bingol and Sheng, 2011). An important
INTRODUCTION means for removing damaged organelles and degrading long-
lived or aggregate-prone proteins is macroautophagy (auto-
Autism spectrum disorders (ASDs) are characterized by im- phagy hereafter), a process downstream of mTOR signaling
paired social interactions, communication deficits, and repetitive that involves the formation of autophagosomes to capture and
behaviors. Multiple ASD susceptibility genes converge on transport cytoplasmic components to lysosomes. The activa-
cellular pathways that intersect at the postsynaptic site of gluta- tion of mTOR inhibits autophagy at an early step in autopha-
matergic synapses (Bourgeron, 2009; Peça and Feng, 2012), gosome formation (Kim et al., 2011). In support of a role for
implicating abnormalities in dendritic spines in ASD pathogen- autophagy dysregulation in ASD etiology, a recent study identi-
esis. Consistently, increased spine density is observed in frontal, fied ASD-associated exonic copy number variation mutations
temporal, and parietal lobes in ASD brains (Hutsler and Zhang, in genes coding for proteins involved in autophagic pathways
2010) and changes in synaptic structure are detected in multiple (Poultney et al., 2013).

Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc. 1131


Neuron
Autism and Neuronal Autophagy

Autophagy has been implicated in synaptic remodeling in revealed profound effects of both disease and age on spine den-
C. elegans (Rowland et al., 2006) and Drosophila (Shen and Ga- sity (p < 0.001, two-way ANOVA, effect of disease: F (1, 16) =
netzky, 2009), but a role in mammalian synaptic development is 73.11, p < 0.001; effect of age: F (1, 16) = 145.7, p < 0.001; dis-
unexplored. We hypothesized that autophagy remodels syn- ease 3 age interaction: F (1, 16) = 28.35, p < 0.001). The spine
apse maturation downstream of mTOR, and autophagy defi- density was slightly higher in childhood ASD patients than con-
ciency downstream of overactivated mTOR contributes to ASD trols (mean ± SD: 12.32 ± 0.60 spines/10 mm in ASD cases versus
synaptic pathology. We found a higher spine density in basal 11.37 ± 0.68 spines/10 mm in controls) but markedly higher
dendrites of layer V pyramidal neurons in ASD patients than in in adolescent ASD patients than controls (10.33 ± 0.74 spines/
controls. The increased spine density was associated with a 10 mm in ASD cases versus 6.24 ± 0.59 spines/10 mm in
defect in net postnatal spine pruning that was correlated with controls). From childhood through adolescence, dendritic spines
hyperactivated mTOR and impaired autophagy. Using Tsc1/2 decreased by 45% in control subjects but only by 16% in
mutant ASD mice and Atg7CKO neuronal autophagy-deficient ASD patients (Figure 1E), demonstrating a developmental defect
mice, we found that aberrant autophagy and mTOR hyperactiva- in net spine pruning in ASD.
tion underlies ASD-like synaptic pathology and correcting auto-
phagy signaling could normalize developmental dendritic spine Disturbed mTOR-Autophagy Signaling and Spine
pruning defects and social behaviors. Pruning in ASD
To test the hypothesis that mTOR-autophagy signaling is
RESULTS disturbed in ASD and associated with ASD spine pathology,
we performed western blot analysis of phospho-mTOR
Dendritic Spine Pruning Deficits in ASD Human Brain (p-mTOR), total mTOR (t-mTOR), phospho-S6 (p-S6), total S6
We measured dendritic spines of basal dendrites of layer V pyra- (t-S6), and the autophagosome marker LC3 and p62 (Figures
midal neurons in the superior middle temporal lobe, Brodmann 2A and 2F) in frozen BA21 brain samples from age-, gender-,
Area 21 (BA21), a region implicated in ASD due to its partici- and PMI- matched ASD patients and controls (demographic
pation in brain networks involved in social and communicative data in Table S3). To determine the relationship between
processes, including language, social and speech perception, mTOR activity and density of dendritic spines, we examined
auditory and visual processing, and comprehension of intentions the protein levels of postsynaptic marker PSD95 and the presyn-
(Redcay, 2008; Zahn et al., 2007). Abnormalities in ASD temporal aptic protein synapsin I (Figure 2A).
lobe have been confirmed by functional imaging and patholog- No effects of PMI, cause of death, brain pH, or length of stor-
ical studies, including disturbed gene transcription profiles (Gar- age on protein levels were detected (Table S2). We observed
bett et al., 2008; Voineagu et al., 2011), increased dendritic spine a decrease in p-mTOR level with age in controls and higher
densities in pyramidal neurons (Hutsler and Zhang, 2010), and p-mTOR level in ASD patients than in controls at 13–20 years
reduced functional specialization (Shih et al., 2011). (Figure 2B). We observed similar changes in p-S6, a reporter
We compared dendritic spine morphology in ASD patients for mTOR activity (Figure 2C). PSD95 protein level was higher
and controls (demographic data in Table S1 available online) in controls aged 2–10 years than controls aged 13–20 years,
using the Golgi-Kopsch technique. In the adolescent group, consistent with normal developmental spine pruning. This
only males were examined to exclude effects of hormone status. decrease in PSD95 level with age was absent in ASD patients
No correlation was revealed between spine density and poten- (Figure 2D), consistent with the lower spine pruning in ASD pa-
tial confounding factors, including postmortem interval (PMI), tients. The presynaptic marker synapsin I exhibited a near-signif-
seizure history, cause of death, brain pH, or tissue storage (Table icant decrease with age in controls but not in patients (Figure 2E).
S2). As in previous studies (Harris et al., 1992), dendritic protru- Levels of p-mTOR were correlated with PSD95 (Figure 2I), indi-
sions with the ratio of head/neck diameter >1 were classified as cating that lower mTOR activity is associated with a higher den-
spines. The spines were characterized by a neck 0.9–3.0 mm dritic spine density in children and adolescents.
long and a spine head diameter of 0.5–2.0 mm (Figures S1A To determine whether impaired autophagy underlies the spine
and S1B). The average spine head diameter (p = 0.519) and spine pruning deficit in human ASD, we characterized basal autophagy
length (p = 0.819) from individual neurons were similar in ASD in postmortem tissue of the temporal lobe in patients with ASD.
patients and controls at all ages examined (Figures S1C and The level of LC3-II, a biomarker that indicates the abundance of
S1D). The mean net spine density per individual was significantly autophagosomes, was significantly lower in ASD patients than
higher in ASD patients than in controls (Figures 1A and 1B: controls throughout childhood and adolescence (Figures 2F
mean ± SD: 11.32 ± 1.23 spines/10 mm versus 8.81 ± 2.77 and 2G), while the level of p62, a protein substrate for autophagy,
spines/10 mm, p = 0.017, two tailed t test). was higher in both childhood and adolescent ASD patients than
Linear regression of spine density with age indicated a sub- controls (Figures 2F and 2H). These data suggest a low level of
stantially greater level of net spine pruning in controls (slope = basal autophagy in ASD temporal cortex throughout develop-
0.40 spines/10 mm/year, R2 = 0.93) than in ASD patients ment. LC3-II and p62 protein levels were not correlated with
(slope = 0.19/10 mm/year, R2 = 0.55; difference from linear any confounding factor for tissue preservation but were corre-
regression of controls, F = 9.4, p = 0.007) (Figure 1C). Due to lated with seizure activity (Table S2), a common feature of ASD.
the limited number of brain samples available, we grouped pa- The impairment in autophagy in ASD patients was confirmed
tients and controls into two age categories: childhood (2–9 years) by immunolabel of LC3-positive puncta in BA21 layer V: the
and adolescence (13–20 years) (Figures 1D and 1E). Analysis cellular area occupied by LC3 puncta and the integrated

1132 Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc.


Neuron
Autism and Neuronal Autophagy

Figure 1. Dendritic Spine Pruning in Temporal Lobe of ASD Patients and Controls
(A) Representative Golgi images for postmortem human temporal lobe (left, 103, stitched from nine separate image stacks), layer V pyramidal neurons with basal
dendritic tree (top middle, 203, pseudocolored in red; bottom middle, 403, pseudocolored in green; scale bar, 50 mm). The right four panels (1003; scale bar,
5 mm) are representative images of proximal basal dendritic segments from two control subjects (C, aged 8 years and 18 years) and two ASD cases (A, aged
7 years and 15 years).
(B) Distribution of spine density (mean ± SD) in basal dendrites after the first bifurcation. Age and diagnosis are indicated for each sample. Controls aged 2–8 years
[C(2–8 years)]: n = 5; controls aged 13–18 years [C(13–18 years)]: n = 5; ASD cases aged 2–8 years [A(2–8 years)]: n = 5; ASD cases aged 13–18 years
[A(13–18 years)]: n = 5. Each point represents the average spine density for each individual neuron measured from each individual.
(C) A linear regression of spine density with age in the control subjects (n = 10) and ASD patients (n = 10). The number of spines per 10 mm was plotted against the
age of each individual. Broken lines indicate 95% confidence intervals.
(D) Spine density (mean ± SD) for the controls and ASD patients in childhood and adolescence. Each point represents the mean spine density for an individual.
Two-way ANOVA, Bonferroni post hoc test. ***p < 0.001, *p < 0.05.
(E) The decrease of spine density with age was greater in the controls than the ASD patients (mean ± SD). ***p < 0.001.

intensity of LC3 puncta were lower in pyramidal neurons from thology in ASD animal models. We focused on mutations in genes
ASD patients than age-matched controls during both early child- encoding tuberous sclerosis complexes TSC1 (hamartin) and
hood and adolescence (Figures S1E, S1F, and S1G). Decreased TSC2 (tuberin), proteins that form a heterodimer that constitutively
autophagy in cortical neurons was confirmed by the accumula- inhibits Rheb to inactivate mTOR (Ehninger and Silva, 2011).
tion of autophagy substrates p62 and ubiquitin (Ub) (Figures Mutations in both Tsc1 and Tsc2 cause mTOR hyperactivation
S1H, S1I, and S1J) and is thus an early feature of ASD. Higher and ASD-like behaviors in mice (Chévere-Torres et al., 2012;
levels of p-mTOR in both control and patients with ASD were Tsai et al., 2012a; Goorden et al., 2007; Sato et al., 2012).
strongly associated with lower levels of LC3-II (Figure 2J), sug- Tsc2+/ mice however exhibit normal social preference in a
gesting that the lower level of autophagy in ASD patients was three-chamber test (Ehninger et al., 2008; Ehninger et al., 2012)
attributable to high mTOR activity. Higher LC3-II levels were but deficient social interaction in a dyadic reciprocal social test
strongly associated with lower levels of PSD95 (Figure 2K). (Sato et al., 2012). This discrepancy in social behaviors could
These data indicate that mTOR-dependent autophagy is nega- be due to differences in testing protocols, the gender, the
tively correlated with spine density in human brain during child- age, or genetic background of the testing mice. We thus char-
hood and adolescence. acterized ASD-like social behaviors in P30–P35 adolescent
male Tsc2+/ mice maintained in a B6/C57 background. We
mTOR Dysregulation Causes Spine Pruning Defects in observed no motor defects or anxiety-like behaviors in open
TSC-Deficient Mouse Models of ASD field (Figures S2A–S2F). In the novel object recognition test,
We then investigated whether mTOR hyperactivation and result- Tsc2+/ mice spent less time exploring the novel object than
ing inhibition of autophagy causes ASD-like dendritic spine pa- their wild-type (WT) littermates, with no difference in time spent

Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc. 1133


Neuron
Autism and Neuronal Autophagy

Figure 2. Dysregulated mTOR-Autophagy Signaling and Spine Pruning in ASD Temporal Lobe
(A) Representative western blots of p-mTOR, t-mTOR, p-S6, t-S6, PSD95, and synapsin I in temporal lobe of ASD patients and control subjects aged 2–9 years
(ASD, n = 8; controls, n = 7) and 13–19 years (ASD, n = 5; controls, n = 9). A, ASD patients; C, controls.
(B–E) The relative density (mean ± SD) for p-mTOR (B) and p-S6 (C) were normalized to t-mTOR and t-S6, respectively. PSD95 (D) and synapsin I (E) levels were
normalized to actin and are presented as scatterplots for ASD patients and controls in two age groups. Each point represents each individual subject. **p < 0.01;
***p < 0.001 (two-way ANOVA, Bonferroni’s post hoc test).
(F) Western blot of autophagy markers, LC3-II and p62, in temporal lobe of ASD patients and control subjects aged 2–9 years and 13–20 years.
(G) LC3-II levels normalized to actin in controls and patients. **p < 0.01; ***p < 0.001 (two-way ANOVA, Bonferroni post hoc test). Mean ± SD.
(H) p62 levels normalized to actin in controls and patients. **p < 0.01; ***p < 0.001 (two-way ANOVA, Bonferroni post hoc test). Mean ± SD.
(I) Correlation between p-mTOR and PSD95 (R2 = 0.598, p < 0.001).
(J) Correlation between p-mTOR and LC3-II in individuals younger than 10 years (R2 = 0.347, p < 0.0001), indicating that LC3-II is regulated by mTOR in both ASD
patients and controls.
(K) Correlation between LC3-II and PSD95 in individuals younger than 10 years (R2 = 0.422, p < 0.0001), suggesting a relationship between synaptic structure
protein levels and autophagy.

exploring the familiar object (Figure 3A). Tsc2+/ mice, how- (Figure 3D, right). In the social novelty test, Tsc2+/ mice spent
ever, did not exhibit ASD-like repetitive behaviors (Figure 3B). a similar amount of time sniffing both novel and familiar social
Sociability was assessed during a dyadic social interaction targets (Figure 3E, left), with decreased preference index (the
with a novel (noncagemate) mouse matched for sex and geno- ratio of time sniffing a stranger mouse versus a familiar mouse;
type (see Supplemental Information). Tsc2+/ mice spent less Figure 3E, right), indicating a reduced preference for social
time sniffing the stimulus mouse (Figure 3C), indicating novelty.
impaired social interactions. Social deficits were confirmed us- The density of dendritic spines in pyramidal neuron basal den-
ing a three-chamber social test. While Tsc2+/ mice showed a drites of layer V A1/S2 in temporal cortex, which is thought to be
preference for interacting with a social target compared with analogous to the primate primary auditory cortex (A1) and sec-
nonsocial target (Figure 3D, left), the preference index (the ratio ondary somatosensory cortex (S2) (Benavides-Piccione et al.,
of time sniffing mouse versus nonsocial target) was decreased 2002), was examined by DiOlistic labeling (Figure 3F). A higher

1134 Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc.


Neuron
Autism and Neuronal Autophagy

Figure 3. Spine Pruning Defects in Tsc1/2-Deficient Cortical Projection Neurons


(A–D) Social behaviors in P30–P33 male adolescent TSC2+/ mice. Tsc2+/+ WT: n = 15, Tsc2+/ : n = 14. Mean ± SEM. (A) Novel object recognition test showing
time spent investigating a familiar versus novel object. (B) ASD-like repetitive behavior. (C) Dyadic reciprocal social interaction test showing time spent sniffing a
stimulus mouse. (D) Sociability in the three-chamber test showing time spent (left) and preference (right) for sniffing a stimulus mouse or an object.
(E) Social novelty in the three-chamber test showing time spent (left) and preference (right) for sniffing a stranger mouse versus a familiar mouse. Compared to WT,
**p < 0.01; *p < 0.05 (unpaired t test). Mean ± SEM.
(F) A confocal image of a DiI-labeled layer V cortical pyramidal neuron. Scale, 20 mm.
(G) Typical confocal images of DiI labeled dendrites in WT, Tsc2+/ and rapamycin (Rapa) treated Tsc2+/ mice at P19–P20 and P29–P30. Rapamycin was
administered at 3 mg/kg/day intraperitoneally from P21 to P28, and the mice were labeled for spine analysis on P29–P30. Scale bar, 2 mm.
(H) Spine pruning in Tsc2+/ mice. ** compared with WT at P29-30, p < 0.01 (two-way ANOVA, Bonferroni post hoc test). n = 7–10 mice per group. Mean ± SD.
(I) Representative images of DiI-labeled dendrites from Tsc1 CKO mutants and Tsc1flox/flox controls. Scale bar, 2 mm.
(J) Spine density in Tsc1flox/flox and Tsc1 CKO mice at P19–P20 and P29–P30. ** compared to P29–P30 Tsc1flox/flox controls, p < 0.01 (two-way ANOVA, Bonferroni
post hoc test). Mean ± SD.

spine density was found in adolescent Tsc2+/ mice than in and basal dendritic tree complexity were similar in WT and
WT (Figures 3G and 3H). The greater spine density in Tsc2+/ Tsc2+/ mutants (Figure S3A). These results suggest that there
layer V cortex at P30 was confirmed by increased immunolabel is a period of massive spine pruning between the ages of P19–
for the presynaptic marker, synaptophysin, and the postsynaptic P20 and P29–P30 in WT but a lack of normal pruning in Tsc2+/
marker, PSD95 (Figures S2G and S2H). We also observed mice. Inhibition of mTOR by intraperitoneal (i.p.) injection of rapa-
increased PSD95 and F-actin-labeled puncta along the den- mycin showed no effects in WT controls (Figure S3B) but cor-
drites of mature Tsc2+/ primary neuronal cultures (Figures rected the pruning defect in Tsc2+/ mutants to the control level.
S2I and S2J). To confirm the effect of the Tsc deficiency on dendritic spine
Net spine pruning normally occurs in mice after the third post- pruning, we used a Tsc1 conditional knockout mouse line
natal week (Zuo et al., 2005), and so we compared spine densities (Tsc1CKO), in which the Tsc1 gene was depleted from pyramidal
between P19–P20 and P29–P30. If the lack of TSC and neurons in the forebrain by crossing Tsc1flox/flox mice to CamKII-
hyperactivation of mTOR led to spine overgrowth, an increase Cre mice. As CamKII promoter-driven Cre recombination begins
in spine density would be expected prior to spine pruning. in layer II-III cortical pyramidal neurons at P19–P20 but is sub-
DiOlistic labeling revealed similar numbers of spines at P19– stantial in all cortical layers at P23–P30 (Figure S4A), the level
P20 in Tsc2+/ and WT mice but far more spines in P29–P30 of TSC1 is normal in deep cortical layers at the start of this
Tsc2+/ mice than in WT (Figures 3G and 3H). The soma size time window for spine pruning and depleted thereafter (Figures

Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc. 1135


Neuron
Autism and Neuronal Autophagy

Figure 4. TSC Ablation Downregulates Autophagic Activity and Rapamycin Reconstitutes Normal Autophagy
(A) Western blot analysis of p-mTOR, t-mTOR, and LC3-II in P29 Tsc2+/ mouse brain. Tsc2 WT and Tsc2+/ mice were i.p. injected with DMSO vehicle or
rapamycin from P20 to P28. Right: quantification of p-mTOR and LC3-II levels. Mean ± SD. *p < 0.05; **p < 0.01; ***p < 0.001 (two-way ANOVA, Bonferroni post
hoc test). n = 5–6 animals per group. Tsc2+/ mutant cortex on P29–P30 showed increased p-mTOR levels and decreased levels of LC3-II. Inhibiting mTOR with
rapamycin decreased p-mTOR and increased LC3II in both wild-type and mutant lines.
(B) Impaired autophagic flux in Tsc2+/ ; GFP-LC3 cortical neurons. Right: mean number of GFP-LC3 puncta per soma of cortical neurons; 8–10 neurons per
group in triplicates were analyzed. Scale bar, 10 mm. *p < 0.05; **p < 0.01; ***p < 0.001 (two-way ANOVA, Bonferroni post hoc test). Mean ± SD.

S4B and S4C). This mouse line allows us to model an appropriate (2) an increase in the level of phospho-S6 (pS6), indicating mTOR
developmental period for evaluating roles for cell-autonomous hyperactivation, and a reduction in GFP-LC3 puncta, a fluores-
effects of neuronal mTOR in the regulation of developmental cent marker for autophagosomes, in cortical pyramidal neurons
spine pruning. At P19–P20, the density of spines in basal den- from Tsc2+/ : GFP-LC3 mice (Figure S5A), indicating impaired
drites of A1/S2 layer V pyramidal neurons was equivalent for autophagy and rapamycin-normalized pS6 levels and numbers
both Tsc1flox/flox control and Tsc1CKO mice (p > 0.05), but spines of GFP-LC3 puncta; (3) decreased immunolabel for endogenous
in control mice were significantly less dense than in Tsc1CKO LC3 in Tsc2+/ mutant primary neuronal cultures (Figure S5B);
mice at P29–P30 (p < 0.01, Figures 3I and 3J). Although soma (4) an accumulation of autophagy substrates, including p62 (Ko-
size was slightly greater (15%) in P29–P30 Tsc1CKO pyramidal matsu et al., 2007a), lipid droplets, and damaged mitochondria
neurons, the number of primary basal dendrites was similar for (Martinez-Vicente et al., 2010), in Tsc2+/ primary neuronal cul-
control and Tsc1CKO mice (Figures S4D, S4E, and S4F). Rapa- tures (Figures S5B, S5C, S5D, and S5E); (5) accumulation of p62-
mycin treatment did not affect spine density in Tsc1flox/flox control and Ub- positive inclusions in pyramidal neurons in Tsc1CKO
mice (Figure S5B), but corrected spine pruning in Tsc1CKO mice mouse brain (Figure S5F).
to control levels (Figures 3I and 3J), with no effect on basal We analyzed autophagy flux in Tsc2+/ :GFP-LC3 neurons
dendritic branching. Thus, both Tsc1CKO and Tsc2+/ mutants in comparison to Tsc2 WT: GFP-LC3 neurons (Figure 4B).
showed a lack of efficient postnatal spine pruning, indicating Cultured primary neurons were treated with bafilomycin
that TSC inhibition of mTOR is required for postnatal spine prun- (BafA1) and NH4Cl to inhibit lysosomal hydrolase and block au-
ing. The effects of TSC1 deletion on spine density in pyramidal tophagosome-lysosome fusion. We reasoned that if autophagy
neurons at P30 is consistent with those reported in vivo in Pur- flux was impaired by mTOR hyperactivation, BafA1/NH4CL
kinje cells (Tsai et al., 2012a). blockade of lysosomal degradation would produce a lower
accumulation of autophagosomes in Tsc2+/ neurons than in
Autophagy Deficiency in Tsc2+/– Mutant Neurons WT. As expected, there was more accumulation of GFP-LC3
We then addressed whether autophagy remodels dendritic puncta in BafA1/NH4CL-treated WT neurons than Tsc2+/
spines downstream of mTOR. We confirmed suppression of neurons, confirming a failure of autophagosome induction
basal autophagy due to mTOR disinhibition in the Tsc mutant in Tsc2+/ neurons (Figure 4B). Rapamycin (200 nM, 8 hr)
mouse brain by (1) a decrease in protein levels of LC3-II in normalized autophagosome formation in Tsc2+/ neurons.
Tsc2+/ cortex, which was normalized by rapamycin (Figure 4A); These findings support the hypothesis that basal neuronal

1136 Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc.


Neuron
Autism and Neuronal Autophagy

Figure 5. Dendritic Spine Pruning Defects and ASD-like Behaviors in Atg7CKO Mice
(A) Western blot analysis of autophagy markers in the Atg7CKO cortex. Brain homogenates from P19–P20 and P29–P30 mice were immunoblotted with antibodies
against Atg12-Atg5, LC3, and autophagy substrate p62. Data shown are representative of three separate experiments. Loss of autophagy was indicated by a
decrease in levels of Atg5-12 conjugation and LC3-II protein, and an increase in p62 protein.
(B) Immunofluorescent labeling of p62 and ubiquitin (Ub) in P30 Atg7CKO mouse cortex. Scale bar, 10 mm.
(C–F) ASD-like social behaviors in Atg7CKO mice. Atg7flox/flox males, n = 15; Atg7CKO males, n = 13. (C) Novel object recognition test showing time spent sniffing a
familiar object versus a novel object. (D) Dyadic social interaction test showing the time testing mice spent sniffing a stimulus mouse. ** Compared to Atg7flox/flox;
p < 0.01; unpaired t test. (E) Sociability in the three-chamber test showing time spent (left) and preference (right) for a stimulus mouse or an object. (F) Social
novelty in the three-chamber test showing time spent (left) and preference (right) for sniffing a stranger mouse versus a familiar mouse. Compared to WT,
**p < 0.01 (unpaired t test). Mean ± SEM.
(G) Dendritic segments from Atg7flox/flox and Atg7CKO pyramidal neurons at P19–P20 and P29–P30. n = 7–10 animals per group. Scale bar, 2 mm.
(H) Fewer spines were pruned in Atg7CKO mice. ** Compared to P29–P30 Atg7flox/flox, p < 0.01 (two-way ANOVA, Bonferroni post hoc test). Mean ± SD.
(I) Timeline of infection and spine analysis.
(J) Cultured control and Atg7 siRNA lentiviral infected mouse hippocampal neurons at DIV20, visualized by GFP and PSD95 fluorescence. Atg7 siRNA expressing
neurons exhibited more PSD95 puncta than controls. Scale bar, 20 mm.
(K) Spine formation and elimination in control and Atg7 siRNA-infected neurons during a 12 hr time window at DIV19–DIV20. Mean ± SD. Scale bar, 10 mm.

autophagy is depressed due to mTOR hyperactivation in Tsc (Ub) (Figures 5B and 6A), proteins that form aggregates after
mutant ASD mouse models. autophagy inhibition (Komatsu et al., 2006, 2007a). At P20
there were no differences in LC3-II and conjugated ATG5-12
Autophagy Deficiency Results in ASD-like Social levels between genotypes, although p62 levels were higher
Behaviors and Spine Pruning Defects in Atg7flox/flox:CamKII-Cre (Atg7CKO) autophagy-deficient mice
To investigate whether neuronal autophagy deficiency produces (Figure 5A). By P30, however, Atg7CKO mice exhibited less con-
ASD-like behaviors and dendritic spine pathology, we generated jugated ATG5-12 and LC3-II protein and more p62 protein than
forebrain excitatory neuronal specific autophagy-deficient mice the Atg7flox/flox controls. Atg7CKO mice displayed occasional
by crossing Atg7flox/flox mice to CamKIIa-cre mice. Atg7 is an p62/Ub-positive inclusions in pyramidal neurons at P20 and
E1-like activating enzyme required for autophagosome forma- prominent p62/Ub-positive aggregates in layer II-III and layer
tion (Komatsu et al., 2006). A deficit in autophagy was confirmed V-VI pyramidal neurons at P30 (Figure 5B). The results confirm
by western blot analysis of conjugated ATG5-12, p62, and LC3-II a loss of autophagy between P20 and P30 in cortical pyramidal
proteins (Figure 5A) and by immunolabel of p62 and ubiquitin neurons in Atg7CKO mice.

Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc. 1137


Neuron
Autism and Neuronal Autophagy

Atg7CKO mice exhibited ASD-like social recognition (Figure 5C) downstream biological processes including protein synthesis,
and social interaction deficits similar to those seen in Tsc2+/ autophagy, ribosome biogenesis, and activation of transcription
mutants. During dyadic encounters, they spent less time sniffing leading to lysosome biogenesis or mitochondrial metabolism.
stimulus mice than their control littermates (Figure 5D). In the To disentangle autophagy from other downstream effectors of
three-chamber test, Atg7CKO mice displayed impaired prefer- mTOR, we crossed Tsc2+/ mice to the Atg7CKO mice to pro-
ence for sniffing the social target (Figure 5E) and for social nov- duce a Tsc2+/ :Atg7CKO double mutant line. We hypothesized
elty (Figure 5F). However, these mice did not show stereotyped that if neuronal autophagy were responsible for spine pruning,
repetitive behaviors, motor defects, or anxiety-like behaviors rapamycin treatment during the fourth week would reconstitute
(Figures S6A–S6G). normal autophagy and pruning in the Tsc2+/ mice but would
We did not observe significant changes in the size of neuronal not do so in Atg7CKO and Tsc2+/ :Atg7CKO double mutant mice.
soma and the number of primary basal dendrites during this We observed high levels of p62 and ubiquitin in Tsc2+/ ,
developmental period (Figures S6H–S6J). At P19–P20, Atg7CKO Atg7CKO, and Tsc2+/ :Atg7CKO double mutant cortices at P30,
basal dendrites from layer V A1/S2 pyramidal neurons exhibited consistent with a deficit of autophagy between P20 and P30 (Fig-
a similar number of spines as those in the Atg7flox/flox control ure 6A). We imaged basal dendrites of layer V A1/S2 pyramidal
mice (Figures 5G and 5H). In contrast, by P29–P30 the Atg7CKO neurons in all lines at P20 to provide a baseline. We then treated
basal dendrites exhibited more spines than controls. Thus, basal mice from all lines with DMSO vehicle or rapamycin from P21 to
neuronal autophagy is required for normal spine pruning during P28. On P29, basal dendrites of layer V A1/S2 pyramidal neurons
postnatal development and for the development of normal social were labeled and analyzed (Figures 6B and 6C). In the DMSO
behaviors. The pruning defect is unlikely to result from abnormal- vehicle-treated mice, the percentage of spines pruned between
ities in microglia and astrocytes, as we observed no activation of P21 and P29 was 26%; 8%, 3%, and 2% were pruned in the
microglia or astrocytes at any age in Atg7CKO mice (Figures S6K Atg7CKO, Tsc2+/ , and double mutant mouse lines, respec-
and S6L). tively, all of which were treated with DMSO vehicle (Figure 6D,
two-way ANOVA, Bonferroni’s post hoc test; genotype 3 treat-
Autophagy Mediates Spine Elimination in Primary ment interaction: F(3,16) = 15.38, p < 0.001; effect of treatment:
Cultures of Hippocampal Neurons F(1,16) = 32.16, p < 0.001; effect of genotype: F(3,16) = 56.17, p <
The increased spine density and reduced spine pruning in 0.001). Therefore, basal levels of autophagy appeared respon-
Atg7CKO mice may result from an increase in synapse formation sible for 70% ([26 8)/26]) of postnatal spine pruning in control
or a decrease in synapse elimination. Primary cultures of hippo- mice. No significant effect of rapamycin on spine pruning in
campal neurons have been used as an in vitro system to investi- Atg7flox/flox control mice was observed. Rapamycin reversed
gate the formation, maturation, and pruning of dendritic spines, spine pruning defects in Tsc2+/ mice to the level of control
in which dendritic spines are formed and pruned during a devel- mice but did not rescue spine pruning in Atg7CKO mice or in
opmental period similar to that in vivo in mouse brain (Orefice Tsc2+/ :Atg7CKO double mutants, demonstrating that neuronal
et al., 2013; Ko et al., 2011; Papa et al., 1995), with spine density autophagy is required for spine elimination in Tsc2+/ mice.
increases between 6–10 days in vitro (DIV6–DIV10), peaks at Note that a relatively small fraction (8%/26% = 30%) of spine
DIV14–DIV21, and decreases after weeks 3 or 4 in vitro in pruning was preserved in Atg7CKO mice, indicating that addi-
neuronal cultures. We thus infected CA1 hippocampal neurons tional mechanisms independent from neuronal autophagy are
with a lentivirus expressing EGFP-Atg7 siRNA or an EGFP con- responsible for the remainder of spine elimination during post-
trol virus at DIV14–DIV15, a period of active synapse formation natal development. Consistently, we found that rapamycin
and stabilization in cultures. Three to four days after infection, rescued spine pruning in Tsc2+/ : Atg7CKO mice to this relatively
the cultures were fixed and stained for postsynaptic marker low level of neuronal autophagy-independent pruning.
PSD95 (Figures 5I and 5J). Neurons expressing Atg7 siRNA
exhibited a higher level of PSD95 puncta at DIV19–DIV20 than Rapamycin Normalizes Social Deficits in Tsc2+/– Mice
neurons infected with control virus (Figure 5J), suggesting an but Not in Atg7 Conditional Knockouts
increased spine density. We calculated the rate of spine genesis We next examined whether rapamycin rescued social deficits in
and spine pruning at DIV19–DIV20 during a 12 hr time window. In Tsc2+/ mice, Atg7CKO mice, and the double mutants. Sociabil-
control neurons infected with viral vector controls, 12% of ity and social novelty were tested with the three-chamber testing
spines were formed and 13% eliminated, indicating equivalent paradigm as above. DMSO vehicle produced no effect on any
rates of spine formation and elimination that reflect stabilized mouse line (Figures 3, 5, and 6). Atg7flox/flox control mice treated
spine densities (Figure 5K). In contrast, 12% of spines were with vehicle preferred to sniff the novel mouse more than the
formed, but only 5% of pre-existing spines were pruned in nonsocial object in the sociability test (Figures 6E and 6F) and
Atg7 siRNA-infected neurons (p < 0.05, t test). Therefore, Atg7 preferred to sniff the stranger mouse more than the familiar
knockdown produced excessive dendritic spines by inhibiting mouse in the social novelty test (Figures 6G and 6H). In contrast,
elimination but exerted no effect on formation. each mutant mouse line displayed impaired preferences for
sociability and social novelty.
Autophagy Deficiency Underlies Spine Pruning Defects The preferences of the Atg7flox/flox control mice were unaf-
in Tsc2+/– Mice fected by rapamycin. In contrast, rapamycin normalized the
We then asked whether autophagy deficiency underlies spine sociability and social novelty preferences of the Tsc2+/ mice.
pruning defects in Tsc2+/ mice. mTOR regulates a number of Rapamycin, however, did not normalize preferences of either

1138 Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc.


Neuron
Autism and Neuronal Autophagy

Figure 6. Autophagy Deficiency Underlies Spine Pruning Defects and ASD-like Social Deficits in Tsc2+/– Mice
(A) p62- and Ub-positive immunolabeled inclusions in autophagy-deficient neurons. Note that P62+ and Ub+ inclusions were occasionally present in Tsc2+/ ,
Atg7CKO, and Tsc2+/ :Atg7CKO cortex at p20 but appear in a majority of cortical neurons in these three lines at P30. No p62+ and Ub+ inclusions were seen in P30
cortical neurons from Atg7flox/flox control mice. Scale bar, 200 mm.
(B–D) Rapamycin normalized dendritic spine pruning in an autophagy-dependent manner. (B) Representative images of dendrites from different mouse lines
treated with DMSO vehicle or rapamycin. Scale, 2 mm. (C) Graphic representation of dendritic spine densities in each condition. Mean ± SD. (D) Percentage of
spines pruned in control and mutant mouse lines. Percentage change in mean spine density (MSD) between P20 and P29 was calculated as: (MSD (P20) – MSD
(P29)) / MSD (P20) 3 100%. Rapamycin rescued spine pruning deficits in Tsc2+/ mice but not in Atg7CKO and had relatively little effect in Tsc2+/ :Atg7CKO
double mutants. n = 7–10 animals per group. *** Compared to DMSO treated Atg7flox/flox controls, p < 0.001; ### Compared to rapamycin treated Atg7flox/flox
controls, p < 0.001; ♪♪♪ compared to DMSO vehicle controls, p < 0.001 (two-way ANOVA, Bonferroni post hoc test). Mean ± SD.
(E–H) Autophagy deficiency blocks the rescue by rapamycin on ASD-like social behaviors in Tsc2+/ mice. n = 10–14 animals per group. (E and F) Sociability:
DMSO-treated Tsc2+/ , Atg7CKO, and Tsc2+/ :Atg7CKO mice each spent less time sniffing social target versus nonsocial target (E) and exhibited
decreased preference (F) for the social target versus nonsocial target. Rapamycin treatment ameliorated impaired sociability in Tsc2+/ mice, but not in Atg7CKO
and Tsc2+/ :Atg7CKO mice. (G and H) Social novelty: DMSO-treated Tsc2+/ , Atg7CKO, and Tsc2+/ :Atg7CKO mice all spent less time sniffing novel mice during
social novelty test (G) and displayed a decrease in preference for social novelty (H). Rapamyin treatment prevented the loss of preference for social novelty in
Tsc2+/ mice but not in Atg7CKO and Tsc2+/ :Atg7CKO mice. *p < 0.05; **p < 0.01; ***p < 0.001 (two-way ANOVA, Bonferroni post hoc test). Mean ± SEM.

the Atg7CKO mice or the double mutants (sociability test, Fig- p < 0.05; effect of treatment: F(1,85) = 13.02, p < 0.001; effect
ure 6F, p < 0.01, two-way ANOVA, Bonferroni’s post hoc test; of genotype: F(3,85) = 25.64, p < 0.001). Thus, rapamycin
genotype 3 treatment interaction: p > 0.05, effect of treatment: rescued impaired sociability and social novelty preferences
F(1,85) = 4.156, p < 0.05; effect of genotype: F(3,85) = 6.775, of Tsc2+/ mice but did not rescue behaviors in Atg7CKO mice
p < 0.001; social novelty test, Figure 6H, p < 0.01, two-way in which neuronal autophagy cannot be activated. Note that
ANOVA: genotype 3 treatment interaction: F(3,85) = 3.085, rapamycin failed to reverse the impaired social behaviors in

Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc. 1139


Neuron
Autism and Neuronal Autophagy

Tsc2+/ :Atg7CKO double mutants, although spine pruning defi- hood, teenage, or early adult years, such as the acquisition of
cits were partially reversed. executive skills such as reasoning, motivation, judgment, lan-
guage, and abstract thought (Goda and Davis, 2003; Sternberg
DISCUSSION and Powell, 1983). Many children diagnosed with ASD reach
adolescence and adulthood with functional disability in these
Dendritic Spine Pruning Defect in the ASD Brain skills, in addition to social and communication deficits (Seltzer
We assessed spine density across development and confirm an et al., 2004). The extended duration for normal spine pruning in
increase in basal dendrite spine density in layer V pyramidal human brain may provide an opportunity for therapeutic inter-
neurons in ASD temporal lobe. Layer V pyramidal neurons are vention of multiple functional domains associated with ASD after
the major excitatory neurons that form cortical-cortical and the disease is diagnosed.
cortical-subcortical projections. Basal dendrites receive excit- While our study examined a single brain region, spine pruning
atory and inhibitory inputs from local sources, and excitatory during early postnatal development occurs in cerebral cortex,
cell types target this compartment almost exclusively (Spruston, cerebellum, olfactory bulb, and hippocampus (Purves and Licht-
2008). The increase in basal dendrite spine density suggests man, 1980; Shinoda et al., 2010). As ASD-related neuropa-
an enhanced local excitatory connectivity, a feature of ASD thology involves disruptions in connectivity across the brain, it
(Belmonte et al., 2004) proposed to cause failure in differenti- is likely that additional ASD brain regions may feature spine prun-
ating signals from noise, prevent development of normal long- ing defects during different periods of synaptic development.
range cortical-cortical and cortical-subcortical communications, Nevertheless, the disorganization of synaptic connectivity in
and underlie neocortical excitation/inhibition imbalance (Sporns the temporal lobe, a central node in the social brain network
et al., 2000; Gogolla et al., 2009). (Gotts et al., 2012), may compromise function of a network of
Note that while signs of ASD can often be detected at anatomically distinct brain regions that underlie global brain
12–18 months, 82% of ASD diagnoses occur at 4 years or older dysfunction and ASD-like social deficits (Normand et al., 2013;
(http://www.cdc.gov/ncbddd/autism/data.html), and CNS tis- Tsai et al., 2012a).
sues from very young ASD patients are extremely rare. As human
brain samples from ASD patients cannot be identified prior mTOR-Regulated Autophagy and ASD Synaptic
to diagnosis, pathological analysis cannot determine whether Pathology
increased spine density precedes symptoms. We therefore re- The genetic heterogeneity of ASD encourages the identification
lied on correlations among an age range of available pathological of steps that converge on common pathways to produce the
specimens, synaptic density, and biochemical markers for anal- clinical syndrome. Dysregulated mTOR signaling has been iden-
ysis. It is remarkable that the only available brain sample suitable tified in autism, fragile X syndrome, tuberous sclerosis, neurofi-
for morphological study of a very young diagnosed ASD patient bromatosis, and PTEN-mediated macroencephaly (Peça and
(age = 3 years) displayed a synaptic density higher than any con- Feng, 2012; Bourgeron, 2009), each of which features altered
trol subject. We find that a defect in net spine pruning was dendritic spine densities. mTOR inhibitors, including rapamycin
responsible for the abnormally high synaptic densities in child- and its analogs, have been examined in clinical trials for treating
hood and adolescent ASD, an observation confirmed in animal ASD and neuropsychological deficits in children with TSC (Sahin,
models. A variety of results indicate that this deficit is due in large 2012).
part to a loss of mTOR-dependent autophagy in neurons. While We find that ASD brains exhibit both disrupted mTOR
synapse formation outpaces synapse elimination at young ages, signaling and synaptic defects. It is highly unlikely that these
yielding the highest synaptic density in early life, significantly patients possessed TSC mutations, and so our findings suggest
reduced cortical autophagy was also apparent in the youngest that mTOR signaling provides a common convergent mecha-
diagnosed ASD patient (age = 2 years, frozen tissue), as indi- nism in ASD. mTOR signaling, however, contributes to protein
cated by low levels of the autophagic vacuole marker LC3-II synthesis required for neuronal survival, development, synaptic
and increased level of autophagy substrates p62. An ongoing plasticity, learning, and memory (Hoeffer and Klann, 2010), and
deficiency in autophagy and impaired spine elimination at prolonged use of mTOR inhibitors may cause adverse effects
younger ages would be expected to increase net spine density (Rodrik-Outmezguine et al., 2011). An important goal is to iden-
and interfere with the dynamic turnover of synapses that orga- tify specific signaling pathways downstream of mTOR that may
nizes neural circuits. Interruption of this maturational organiza- provide more precise targets. For example, a link has been es-
tion of the brain would lead to a persistence of immature or tablished between eIF4E-dependent translational control down-
formation of aberrant circuits in ASD. stream of mTOR and ASD-like phenotypes in mouse models
The near-linear decrease in spine number from all cases be- (Santini et al., 2013). We provide evidence from postmortem
tween the ages of 2 to 19 years indicates that spine pruning in brain that autophagy deficiency, which is a consequence of
temporal lobe occurs over the first two decades and that net mTOR overactivation, strongly correlates with ASD dendritic
loss of synapses is substantially greater in controls than ASD spine pathology. The reduction of mTOR-regulated neuronal
patients. The spine densities declined during the first and second autophagy is further consistent with our recent findings of a
decade by 41% in normal controls but only by 16% in ASD pa- lack of autophagic mitochondrial turnover in ASD brains (Tang
tients, a level independently confirmed by analysis of pre- and et al., 2013).
postsynaptic markers. This deficit may contribute to abnormal- We have confirmed in mouse models that inhibition of
ities in cognitive functions that humans acquire in their late child- neuronal autophagy produced ASD-like inhibition of normal

1140 Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc.


Neuron
Autism and Neuronal Autophagy

developmental spine pruning and ASD-like behaviors. Pharma- pointing to the possibility of glial non-cell-autonomous autopha-
cological inhibition of mTOR activity normalized ASD-like spine gic regulation of spine morphogenesis. In addition, changes in
pruning deficits and ASD-like behaviors in mice largely by mTOR-autophagy signaling and spine pruning defects may
activating neuronal autophagy. As these data suggest a direct represent a secondary mechanism in response to an imbalance
link between mTOR-regulated autophagy and pruning of syn- between excitatory and inhibitory neurotransmission, identified
aptic connections during postnatal development, developing in both Mecp2 mutant mice and Tsc1-deficient mice and impli-
targeted means to enhance autophagy downstream of mTOR cated in ASD-associated stereotypies and social behavioral def-
during development may provide the basis for novel ASD icits (Chao et al., 2010; Fu et al., 2012; Yizhar et al., 2011). Altered
therapeutics. synaptic function is consistent with our recent finding that
chronic lack of neuronal autophagy enhances evoked neuro-
Autophagy and Spine Pruning transmitter release and rate of synaptic recovery (Hernandez
The precise control of synapse pruning could be achieved by et al., 2012).
multiple signaling systems that converge to eliminate synaptic In summary, we find that many ASD brains exhibit both dis-
connections. This could involve the targeted degradation of syn- rupted mTOR signaling and synaptic defects during childhood
aptic components. Recent evidence suggests that neuronal ac- and adolescence, suggesting that mTOR signaling may provide
tivity decreases dendritic spine number in part through activation a common mechanism involved in ASD synaptic pathology
of the myocyte enhancer factor 2 (MEF2) transcription factor (Sawicka and Zukin, 2012). We further demonstrated that ASD
(Pfeiffer et al., 2010), which promotes ubiquitin-proteasome sys- behaviors and synaptic deficits are elicited by altered mTOR
tem (UPS)-dependent degradation of the synaptic scaffolding signaling via an inhibition of autophagy required for normal
protein PSD95 (Tsai et al., 2012b). In addition to the UPS, which developmental spine pruning. The results indicate a direct link
is primarily responsible for the degradation of short-lived cyto- between mTOR-autophagy and pruning of synaptic connec-
solic proteins, neurons rely on lysosomal-dependent degrada- tions during postnatal development and suggest that targeting
tion mechanisms for the turnover of long-lived synaptic proteins neuronal autophagy could provide therapeutic benefit.
and damaged organelles. Ablation of autophagy genes ATG7
or ATG5 causes neurodegeneration associated with aberrant EXPERIMENTAL PROCEDURES
organelles and ubiquitin-rich inclusions in neuronal cell bodies
ASD-like Social Behavioral Tests
(Hara et al., 2006; Komatsu et al., 2006), as well as disrupted
Mice were tested for novel object recognition and social interactions, anxiety-
membrane homeostasis in axon terminals (Komatsu et al.,
like behaviors, exploratory locomotion behaviors, and self-grooming repetitive
2007b; Hernandez et al., 2012). behavior. Sociability and social novelty were tested in a three-chamber testing
Using an in vitro primary neuronal culture system, we find that paradigm. Procedures were approved by Columbia University IACUC.
autophagy regulates spine elimination but not spine formation
during developmental pruning of dendritic spines. Autophagy Biochemistry, DiOlistic Labeling, Golgi Staining, and
may remodel dendritic spines by directing internalized postsyn- Immunohistochemistry
Mouse and human brain tissue were lysed with 1X RIPA buffer supplemented
aptic membrane neurotransmitter receptors, including GABA-A
with protease inhibitors and phosphatase inhibitors, and subjected to western
(Rowland et al., 2006) and AMPAR (Shehata et al., 2012), toward blot analysis. Neurons in mouse brain were labeled with DiI using a Helios gene
lysosomal degradation. Although autophagy was classically gun system at 120 psi. Fluorescent image stacks were acquired with a Leica
considered an ‘‘in-bulk’’ process, evidence now supports selec- multiphoton system. Neuronal morphology in postmortem human brain was
tivity mediated via recognition of posttranslational modifications analyzed by Golgi-Kopsch technique. Images were reconstructed with Imaris
by molecules that bind cargo and components of the autophagic FilamentTracer Module (Bitplane).
Full Methods and associated references are in the Supplemental
machinery. p62 is the most extensively characterized cargo-
Information.
recognizing molecule and binds preferentially to an ubiquitin link-
age (Lys63) on the surface of ubiquitinated protein aggregates,
SUPPLEMENTAL INFORMATION
polyubiquitinated proteins, and organelles. In addition, auto-
phagy may degrade proteins that suppress spine elimination, Supplemental Information includes Supplemental Experimental Procedures,
and the loss of autophagy could accumulate proteins that block six figures, and three tables and can be found with this article online at
spine pruning, for example, by releasing translationally sup- http://dx.doi.org/10.1016/j.neuron.2014.07.040.
pressed synaptic mRNA for local protein synthesis (Banerjee
AUTHOR CONTRIBUTIONS
et al., 2009).
As neuronal autophagy is responsible for 70% of postnatal G.T. and D.S. conceived and designed the study. G.T. and M.L.C. performed
net spine elimination, it is likely that basal autophagy regulates and analyzed DiOlistic labeling experiments; K.G. and F.C. designed, per-
spine elimination in cooperation with additional regulatory mech- formed, and analyzed all behavioral experiments; G.T. and C.B. performed
anisms downstream of mTOR, including eIF4E-dependent trans- mouse breeding; and E.K. made neuronal cultures. G.T. and A.S. performed
lational control and neuronal outgrowth (Santini et al., 2013) and biochemistry, Golgi staining, immunolabeling of mouse and human brains,
and establishing neuronal cultures. S.H.K. and M.S. performed data analysis.
other nonneuronal intrinsic regulatory mechanisms including
G.R., A.J.D., and J.E.G. supervised brain sample selection, Golgi staining,
neuroimmune disturbances and astrocyte activation (Garbett
and data interpretation in human subjects. Z.Y., A.Y., and O.A. assisted
et al., 2008; Voineagu et al., 2011; Paolicelli et al., 2011; Schafer with the design of autophagy and behavioral study. G.T., J.E.G., B.S.P.,
et al., 2012; Chung et al., 2013). Defective neuronal autophagy and D.S. wrote the manuscript. All authors read and approved the final
can be induced by infected microglia (Alirezaei et al., 2008), version.

Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc. 1141


Neuron
Autism and Neuronal Autophagy

ACKNOWLEDGMENTS Goorden, S.M., van Woerden, G.M., van der Weerd, L., Cheadle, J.P., and
Elgersma, Y. (2007). Cognitive deficits in Tsc1+/- mice in the absence of
This study was supported by the Simons Foundation. Additional support for cerebral lesions and seizures. Ann. Neurol. 62, 648–655.
D.S. is from DOD TSCRP (TS110056) and the Parkinson’s Disease and JPB Gotts, S.J., Simmons, W.K., Milbury, L.A., Wallace, G.L., Cox, R.W., and
Foundations, for G.T. from NIMH (K01MH096956), for M.L.C. from AHA, for Martin, A. (2012). Fractionation of social brain circuits in autism spectrum dis-
A.J.D. from NIMH (MH64168), for F.C. from NIH (DP2OD001674-01), for orders. Brain 135, 2711–2725.
O.A. from NIH (NS049442). We thank the Autism Tissue Portal, Harvard Brain
Hara, T., Nakamura, K., Matsui, M., Yamamoto, A., Nakahara, Y., Suzuki-
Bank, and Maryland NICHD Brain & Tissue Bank for kindly providing us brain
Migishima, R., Yokoyama, M., Mishima, K., Saito, I., Okano, H., and
tissues for the present study. We thank Ana Maria Cuervo for reagents and
Mizushima, N. (2006). Suppression of basal autophagy in neural cells causes
valuable advice.
neurodegenerative disease in mice. Nature 441, 885–889.

Accepted: July 24, 2014 Harris, K.M., Jensen, F.E., and Tsao, B. (1992). Three-dimensional structure of
Published: August 21, 2014 dendritic spines and synapses in rat hippocampus (CA1) at postnatal day 15
and adult ages: implications for the maturation of synaptic physiology and
REFERENCES long-term potentiation. J. Neurosci. 12, 2685–2705.
Hernandez, D., Torres, C.A., Setlik, W., Cebrián, C., Mosharov, E.V., Tang, G.,
Alirezaei, M., Kiosses, W.B., Flynn, C.T., Brady, N.R., and Fox, H.S. (2008). Cheng, H.C., Kholodilov, N., Yarygina, O., Burke, R.E., et al. (2012). Regulation
Disruption of neuronal autophagy by infected microglia results in neurodegen- of presynaptic neurotransmission by macroautophagy. Neuron 74, 277–284.
eration. PLoS ONE 3, e2906. Hoeffer, C.A., and Klann, E. (2010). mTOR signaling: at the crossroads of plas-
Banerjee, S., Neveu, P., and Kosik, K.S. (2009). A coordinated local transla- ticity, memory and disease. Trends Neurosci. 33, 67–75.
tional control point at the synapse involving relief from silencing and MOV10 Hutsler, J.J., and Zhang, H. (2010). Increased dendritic spine densities on
degradation. Neuron 64, 871–884. cortical projection neurons in autism spectrum disorders. Brain Res. 1309,
Belmonte, M.K., Allen, G., Beckel-Mitchener, A., Boulanger, L.M., Carper, 83–94.
R.A., and Webb, S.J. (2004). Autism and abnormal development of brain con- Kim, J., Kundu, M., Viollet, B., and Guan, K.L. (2011). AMPK and mTOR regu-
nectivity. J. Neurosci. 24, 9228–9231. late autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 13,
Benavides-Piccione, R., Ballesteros-Yáñez, I., DeFelipe, J., and Yuste, R. 132–141.
(2002). Cortical area and species differences in dendritic spine morphology. Ko, J., Soler-Llavina, G.J., Fuccillo, M.V., Malenka, R.C., and Südhof, T.C.
J. Neurocytol. 31, 337–346. (2011). Neuroligins/LRRTMs prevent activity- and Ca2+/calmodulin-depen-
Bingol, B., and Sheng, M. (2011). Deconstruction for reconstruction: the role of dent synapse elimination in cultured neurons. J. Cell Biol. 194, 323–334.
proteolysis in neural plasticity and disease. Neuron 69, 22–32.
Komatsu, M., Waguri, S., Chiba, T., Murata, S., Iwata, J., Tanida, I., Ueno, T.,
Bourgeron, T. (2009). A synaptic trek to autism. Curr. Opin. Neurobiol. 19, Koike, M., Uchiyama, Y., Kominami, E., and Tanaka, K. (2006). Loss of auto-
231–234. phagy in the central nervous system causes neurodegeneration in mice.
Chao, H.T., Chen, H., Samaco, R.C., Xue, M., Chahrour, M., Yoo, J., Neul, J.L., Nature 441, 880–884.
Gong, S., Lu, H.C., Heintz, N., et al. (2010). Dysfunction in GABA signalling me- Komatsu, M., Waguri, S., Koike, M., Sou, Y.S., Ueno, T., Hara, T., Mizushima,
diates autism-like stereotypies and Rett syndrome phenotypes. Nature 468, N., Iwata, J., Ezaki, J., Murata, S., et al. (2007a). Homeostatic levels of p62
263–269. control cytoplasmic inclusion body formation in autophagy-deficient mice.
Chévere-Torres, I., Maki, J.M., Santini, E., and Klann, E. (2012). Impaired social Cell 131, 1149–1163.
interactions and motor learning skills in tuberous sclerosis complex model Komatsu, M., Wang, Q.J., Holstein, G.R., Friedrich, V.L., Jr., Iwata, J.,
mice expressing a dominant/negative form of tuberin. Neurobiol. Dis. 45, Kominami, E., Chait, B.T., Tanaka, K., and Yue, Z. (2007b). Essential role for
156–164. autophagy protein Atg7 in the maintenance of axonal homeostasis and the
Chung, W.S., Clarke, L.E., Wang, G.X., Stafford, B.K., Sher, A., Chakraborty, prevention of axonal degeneration. Proc. Natl. Acad. Sci. USA 104, 14489–
C., Joung, J., Foo, L.C., Thompson, A., Chen, C., et al. (2013). Astrocytes 14494.
mediate synapse elimination through MEGF10 and MERTK pathways. Martinez-Vicente, M., Talloczy, Z., Wong, E., Tang, G., Koga, H., Kaushik, S.,
Nature 504, 394–400. de Vries, R., Arias, E., Harris, S., Sulzer, D., and Cuervo, A.M. (2010). Cargo
Ehninger, D., and Silva, A.J. (2011). Rapamycin for treating Tuberous sclerosis recognition failure is responsible for inefficient autophagy in Huntington’s dis-
and Autism spectrum disorders. Trends Mol. Med. 17, 78–87. ease. Nat. Neurosci. 13, 567–576.
Ehninger, D., Han, S., Shilyansky, C., Zhou, Y., Li, W., Kwiatkowski, D.J., Normand, E.A., Crandall, S.R., Thorn, C.A., Murphy, E.M., Voelcker, B.,
Ramesh, V., and Silva, A.J. (2008). Reversal of learning deficits in a Tsc2+/- Browning, C., Machan, J.T., Moore, C.I., Connors, B.W., and Zervas, M.
mouse model of tuberous sclerosis. Nat. Med. 14, 843–848. (2013). Temporal and mosaic Tsc1 deletion in the developing thalamus dis-
Ehninger, D., Sano, Y., de Vries, P.J., Dies, K., Franz, D., Geschwind, D.H., rupts thalamocortical circuitry, neural function, and behavior. Neuron 78,
Kaur, M., Lee, Y.S., Li, W., Lowe, J.K., et al. (2012). Gestational immune acti- 895–909.
vation and Tsc2 haploinsufficiency cooperate to disrupt fetal survival and may Orefice, L.L., Waterhouse, E.G., Partridge, J.G., Lalchandani, R.R., Vicini, S.,
perturb social behavior in adult mice. Mol. Psychiatry 17, 62–70. and Xu, B. (2013). Distinct roles for somatically and dendritically synthesized
Fu, C., Cawthon, B., Clinkscales, W., Bruce, A., Winzenburger, P., and Ess, brain-derived neurotrophic factor in morphogenesis of dendritic spines.
K.C. (2012). GABAergic interneuron development and function is modulated J. Neurosci. 33, 11618–11632.
by the Tsc1 gene. Cereb. Cortex 22, 2111–2119. Paolicelli, R.C., Bolasco, G., Pagani, F., Maggi, L., Scianni, M., Panzanelli, P.,
Garbett, K., Ebert, P.J., Mitchell, A., Lintas, C., Manzi, B., Mirnics, K., and Giustetto, M., Ferreira, T.A., Guiducci, E., Dumas, L., et al. (2011). Synaptic
Persico, A.M. (2008). Immune transcriptome alterations in the temporal cortex pruning by microglia is necessary for normal brain development. Science
of subjects with autism. Neurobiol. Dis. 30, 303–311. 333, 1456–1458.
Goda, Y., and Davis, G.W. (2003). Mechanisms of synapse assembly and Papa, M., Bundman, M.C., Greenberger, V., and Segal, M. (1995).
disassembly. Neuron 40, 243–264. Morphological analysis of dendritic spine development in primary cultures of
Gogolla, N., Leblanc, J.J., Quast, K.B., Südhof, T.C., Fagiolini, M., and hippocampal neurons. J. Neurosci. 15, 1–11.
Hensch, T.K. (2009). Common circuit defect of excitatory-inhibitory balance Peça, J., and Feng, G. (2012). Cellular and synaptic network defects in autism.
in mouse models of autism. J. Neurodev. Disord. 1, 172–181. Curr. Opin. Neurobiol. 22, 866–872.

1142 Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc.


Neuron
Autism and Neuronal Autophagy

Penzes, P., Cahill, M.E., Jones, K.A., VanLeeuwen, J.E., and Woolfrey, K.M. is involved in AMPA receptor degradation after chemical long-term depres-
(2011). Dendritic spine pathology in neuropsychiatric disorders. Nat. sion. J. Neurosci. 32, 10413–10422.
Neurosci. 14, 285–293. Shen, W., and Ganetzky, B. (2009). Autophagy promotes synapse develop-
Pfeiffer, B.E., Zang, T., Wilkerson, J.R., Taniguchi, M., Maksimova, M.A., ment in Drosophila. J. Cell Biol. 187, 71–79.
Smith, L.N., Cowan, C.W., and Huber, K.M. (2010). Fragile X mental retardation Shih, P., Keehn, B., Oram, J.K., Leyden, K.M., Keown, C.L., and Müller, R.A.
protein is required for synapse elimination by the activity-dependent transcrip- (2011). Functional differentiation of posterior superior temporal sulcus in
tion factor MEF2. Neuron 66, 191–197. autism: a functional connectivity magnetic resonance imaging study. Biol.
Poultney, C.S., Goldberg, A.P., Drapeau, E., Kou, Y., Harony-Nicolas, H., Psychiatry 70, 270–277.
Kajiwara, Y., De Rubeis, S., Durand, S., Stevens, C., Rehnström, K., et al. Shinoda, Y., Tanaka, T., Tominaga-Yoshino, K., and Ogura, A. (2010).
(2013). Identification of small exonic CNV from whole-exome sequence data Persistent synapse loss induced by repetitive LTD in developing rat hippocam-
and application to autism spectrum disorder. Am. J. Hum. Genet. 93, 607–619. pal neurons. PLoS ONE 5, e10390.
Purves, D., and Lichtman, J.W. (1980). Elimination of synapses in the devel- Sporns, O., Tononi, G., and Edelman, G.M. (2000). Theoretical neuroanatomy:
oping nervous system. Science 210, 153–157. relating anatomical and functional connectivity in graphs and cortical connec-
Rakic, P., Bourgeois, J.P., Eckenhoff, M.F., Zecevic, N., and Goldman-Rakic, tion matrices. Cereb. Cortex 10, 127–141.
P.S. (1986). Concurrent overproduction of synapses in diverse regions of the Spruston, N. (2008). Pyramidal neurons: dendritic structure and synaptic inte-
primate cerebral cortex. Science 232, 232–235. gration. Nat. Rev. Neurosci. 9, 206–221.
Redcay, E. (2008). The superior temporal sulcus performs a common function Sternberg, R.J., and Powell, J.S. (1983). The development of intelligence. In
for social and speech perception: implications for the emergence of autism. Handbook of Child Psychology, 4th Edition, P.H. Mussen, ed. (New York:
Neurosci. Biobehav. Rev. 32, 123–142. John Wiley and Sons), pp. 341–419.
Rodrik-Outmezguine, V.S., Chandarlapaty, S., Pagano, N.C., Poulikakos, P.I., Tang, G., Gutierrez Rios, P., Kuo, S.H., Akman, H.O., Rosoklija, G., Tanji, K.,
Scaltriti, M., Moskatel, E., Baselga, J., Guichard, S., and Rosen, N. (2011). Dwork, A., Schon, E.A., Dimauro, S., Goldman, J., and Sulzer, D. (2013).
mTOR kinase inhibition causes feedback-dependent biphasic regulation of Mitochondrial abnormalities in temporal lobe of autistic brain. Neurobiol. Dis.
AKT signaling. Cancer Discov 1, 248–259. 54, 349–361.

Rowland, A.M., Richmond, J.E., Olsen, J.G., Hall, D.H., and Bamber, B.A. Tsai, P.T., Hull, C., Chu, Y., Greene-Colozzi, E., Sadowski, A.R., Leech, J.M.,
(2006). Presynaptic terminals independently regulate synaptic clustering and Steinberg, J., Crawley, J.N., Regehr, W.G., and Sahin, M. (2012a). Autistic-like
autophagy of GABAA receptors in Caenorhabditis elegans. J. Neurosci. 26, behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant mice. Nature
1711–1720. 488, 647–651.

Sahin, M. (2012). Targeted treatment trials for tuberous sclerosis and autism: Tsai, N.P., Wilkerson, J.R., Guo, W., Maksimova, M.A., DeMartino, G.N.,
no longer a dream. Curr. Opin. Neurobiol. 22, 895–901. Cowan, C.W., and Huber, K.M. (2012b). Multiple autism-linked genes mediate
synapse elimination via proteasomal degradation of a synaptic scaffold PSD-
Santini, E., Huynh, T.N., MacAskill, A.F., Carter, A.G., Pierre, P., Ruggero, D., 95. Cell 151, 1581–1594.
Kaphzan, H., and Klann, E. (2013). Exaggerated translation causes synaptic
Voineagu, I., Wang, X., Johnston, P., Lowe, J.K., Tian, Y., Horvath, S., Mill, J.,
and behavioural aberrations associated with autism. Nature 493, 411–415.
Cantor, R.M., Blencowe, B.J., and Geschwind, D.H. (2011). Transcriptomic
Sato, A., Kasai, S., Kobayashi, T., Takamatsu, Y., Hino, O., Ikeda, K., and analysis of autistic brain reveals convergent molecular pathology. Nature
Mizuguchi, M. (2012). Rapamycin reverses impaired social interaction in 474, 380–384.
mouse models of tuberous sclerosis complex. Nat. Commun. 3, 1292.
Yizhar, O., Fenno, L.E., Prigge, M., Schneider, F., Davidson, T.J., O’Shea, D.J.,
Sawicka, K., and Zukin, R.S. (2012). Dysregulation of mTOR signaling in neuro- Sohal, V.S., Goshen, I., Finkelstein, J., Paz, J.T., et al. (2011). Neocortical exci-
psychiatric disorders: therapeutic implications. Neuropsychopharmacology tation/inhibition balance in information processing and social dysfunction.
37, 305–306. Nature 477, 171–178.
Schafer, D.P., Lehrman, E.K., Kautzman, A.G., Koyama, R., Mardinly, A.R., Zahn, R., Moll, J., Krueger, F., Huey, E.D., Garrido, G., and Grafman, J. (2007).
Yamasaki, R., Ransohoff, R.M., Greenberg, M.E., Barres, B.A., and Stevens, Social concepts are represented in the superior anterior temporal cortex. Proc.
B. (2012). Microglia sculpt postnatal neural circuits in an activity and comple- Natl. Acad. Sci. USA 104, 6430–6435.
ment-dependent manner. Neuron 74, 691–705. Zoghbi, H.Y., and Bear, M.F. (2012). Synaptic dysfunction in neurodevelop-
Seltzer, M.M., Shattuck, P., Abbeduto, L., and Greenberg, J.S. (2004). mental disorders associated with autism and intellectual disabilities. Cold
Trajectory of development in adolescents and adults with autism. Ment. Spring Harb. Perspect. Biol. 4, a009886.
Retard. Dev. Disabil. Res. Rev. 10, 234–247. Zuo, Y., Yang, G., Kwon, E., and Gan, W.B. (2005). Long-term sensory depri-
Shehata, M., Matsumura, H., Okubo-Suzuki, R., Ohkawa, N., and Inokuchi, K. vation prevents dendritic spine loss in primary somatosensory cortex. Nature
(2012). Neuronal stimulation induces autophagy in hippocampal neurons that 436, 261–265.

Neuron 83, 1131–1143, September 3, 2014 ª2014 Elsevier Inc. 1143


Neuron, Volume 83
Supplemental Information

Loss of mTOR-Dependent Macroautophagy


Causes Autistic-like Synaptic Pruning Deficits
Guomei Tang, Kathryn Gudsnuk, Sheng-Han Kuo, Marisa L. Cotrina, Gorazd Rosoklija,
Alexander Sosunov, Mark S. Sonders, Ellen Kanter, Candace Castagna, Ai Yamamoto,
Zhenyu Yue, Ottavio Arancio, Bradley S. Peterson, Frances Champagne, Andrew J.
Dwork, James Goldman, and David Sulzer
Loss of mTOR-dependent macroautophagy causes autistic-like
synaptic pruning deficits

Inventory of Supplemental Information

1) Supplemental Figure S1;

2) Supplemental Figure S2;

3) Supplemental Figure S3;

4) Supplemental Figure S4;

5) Supplemental Figure S5;

6) Supplemental Figure S6;

7) Supplemental Figure Legends;

8) Supplemental Table S1;

9) Supplemental Table S2;

10) Supplemental Table S3;

11) Experimental Procedures;

12) References for Experimental Procedures

 
E
A
Average spine length

I
F
Average spine length

J
(μm)
Integrated intensity Integrated intensity

A1
C1
Figure S1

A2
C2
C3

A3

J
G

K
Integrated intensity Area fraction

A4
C4
B

Average spine head


C5

A5 diameter (μm)
0.0
1.0
2.0

0.5
1.5

C6

A6
H
Control
C

Average head Average length


diameter (μm) (μm)
ASD
C(2-8) C(13-18) A(3-9) A(13-19)

C(2-8) C(13-18) A(3-9) A(13-19)


Figure S2
A B C Tsc2+/+
80 Tsc2+/+ 400 Tsc2+/+ 60

Total distance (m)


Tsc2+/-

in center (s)
Tsc2+/-

immobile (s)
Tsc2+/-

Total ime
300

Total time
40
40 200
20
100

0 0 0
Male Female Male Female Male Female

D 8 E 120
F 30
Tsc2+/+

Time foraging (s)


Time rearing (s)
Tsc2+/-
# fecal Boli

6
80 20
4
40 10
2
0 0 0
Male Female Tsc2+/+ Tsc2+/- Tsc2+/+ Tsc2+/-

G Tsc2+/+ Tsc2+/- H Tsc2+/+ Tsc2+/- I

Tsc2+/+

Tsc2+/-

J
SYNP Tsc2+/+

Tsc2+/-
Figure S3

A Tsc2+/+ Tsc2+/-
1000

Soma size (µm2)

# of branches
500

0
1
Tsc2+/+ 2
Tsc2+/- Tsc2+/+ Tsc2+/-

Tsc2+/+

Tsc2+/+ Tsc1flox/flox
Figure S4

A B Tsc1flox/flox Tsc1CKO

P18
C Tsc1flox/flox Tsc1CKO

P20

P23

D E F
Tsc1flox/flox Tsc1CKO Tsc1CKO; Rapa 10

# of branches
Soma size (µm2)

0
Tsc1flox/flox Tsc1CKO Tsc1CKO; Tsc11 2 CKO Tsc1
flox/flox Tsc1 3 CKO;
Rapa Rapa
D
Fluorecent intensity
MitoRed / mitoGreen
Figure S5

FE
Figure S6
B C D 80 Atg7flox/flox
A

Total distance (m)


1000

Soma size (µm2)


Atg7flox/flox Atg7CKO Atg7CKO

# of branches
60

500 40

20

0 0
1
Atg7flox/flox 2CKO
Atg7 Atg7flox/flox Atg7CKO Male Female

E F G H
500 60 Atg7flox/flox 8
Total ime immobile (s)

Atg7flox/flox 8

Time grooming (s)


Atg7flox/flox

Total ime in center (s)


Atg7CKO Atg7CKO
400 Atg7CKO
6 6

# fecal boli
40
300
4 4
200
20
100 2 2

0 0 0 0
Male Female Male Female Male Female Atg7flox/floxAtg7CKO

I 100 P20 P30 9mo


L
Time rearing (s)

K P20 P30 9mo


75

50
Atg7flox/flox
25 Atg7flox/flox

0
Atg7flox/flox Atg7CKO

J 30
Atg7CKO

Atg7CKO
Time foraging (s)

20

10 Positive
control

0
Atg7flox/flox Atg7CKO
Supplemental Figure legends

Figure S1. Dendritic spine length and spine head diameter and autophagy markers in temporal lobe

layer V pyramidal neurons from ASD patients: related to Figures 1 & 2. (A) Distribution of spine length in

controls and ASD patients. Each circle represents the average spine length for each individual neuron imaged;

(B) Distribution of spine head diameter in controls and ASD patients. Each circle represents the average spine

head diameter for one individual neuron imaged. Mean ± SD was plotted. (C&D) Average spine length and

spine head diameter for control and ASD patients. Each circle represents the average spine density (C) or

spine head diameter (D) for each individual. Controls aged 2-8yrs [C(2-8yrs)]: n=5; controls aged 13-18yrs

[C(13-18yrs)]: n=5; ASD cases aged 2-8yrs [A(2-8yrs)]: n=5; ASD cases aged 13-18yrs [A(13-18yrs)]: n=5.

(E) Representative images for LC3 immunohistochemistry from 6 control cases (C1, 2y; C2, 2y; C3, 4y; C4, 5y;

C5, 8y; C6, 13y) and 6 ASD cases (A1, 3y; A2, 7y; A3, 7y; A4, 7y; A5, 9y, A6, 14y) in temporal lobe layer V

pyramidal neurons (C1-A5: fluorescence; C6 & A6: DAB). (F&G) Quantification of LC3 puncta in layer V

pyramidal neurons in temporal lobe of ASD patients and control subjects aged 2-8 yrs and 13-18yrs; Data are

presented as the mean integrated intensity of LC3 puncta per neuron (F) and the mean cellular area occupied

by LC3 puncta (G). 10-15 neurons from each case per age group were quantified. Compared to control

subjects, ASD patients showed a decrease in LC3 puncta, indicating a loss of autophagic activity. * p<0.05; **

p<0.01; *** p<0.001. (H-J) p62 and ubiquitin immunohistochemistry in ASD and control temporal lobe layer V

pyramidal neurons. Representative fluorescent images from 5 control cases (C1, 2y; C2, 2y; C3, 4y; C4, 5y;

C5, 8y) and 5 ASD cases (A1, 3y; A2, 7y; A3, 7y; A4, 7y; A5, 9y) are presented in (H). The mean integrated

intensity of fluorescence was quantified for p62 (I) or ubiquitin (J) per pyramidal neuron in control and ASD

subjects. 10-15 neurons from each case per age group were analyzed. Compared to controls, * p<0.05; **

p<0.01; *** p<0.001. Each point represents an individual. C(2-8yrs): n=5; C(13-18yrs): n=5; A(2-8yrs): n=5;

A(13-18yrs): n=5.

Figure S2. Open field behaviors and synaptic density in Tsc2+/- mice related to Figure 3. (A-F) Open

field measures of locomotor activity and anxiety-like behavior in Tsc2+/- mice (A) Total distance traversed

(meter, m); (B) time (second, s) spent immobile; (C) time (s) spent in the center arena; (D) number of fecal

 
boli; (E) frequency of rearing during the test session; (F) frequency of digging/foraging during the test session.

No genotype and gender differences were found for all parameters. n (Tsc2+/+, wt, male)=15; n (Tsc2+/+,

female)=20; n (Tsc2+/-, male)=14; n (Tsc2+/-, female)=12. Data are presented as mean ± SEM. (G-J)

Increased synaptic density in Tsc2+/- brain and neuronal cultures. Tsc2+/- mice were crossed to mice

expressing GFP-LC3, a fluorescent membrane marker for autophagosomes. (G&H) Control and Tsc2+/-

cortical sections were immunolabeled with antibodies against postsynaptic marker PSD95, presynaptic marker

synaptophysin (SYNP), and GFP. Compared with Tsc2+/+ wt, Tsc2+/- mouse cortex showed increased PSD95

(G) and SYNP (H) puncta, but decreased GFP-LC3 puncta; (I&J) Postsynaptic marker proteins PSD95 and F-

actin in primary cortical neuronal cultures from wt and Tsc2+/- mice at day 19-20 in vitro. We observed an

increase in PSD95 (I), pS6 (J) and F-actin (J) puncta along Tsc2+/- neurites.

Figure S3. Neuronal morphology in Tsc2+/- mouse brain and effects of rapamycin on dendritic spine

density in control mice, related to Figure 3. (A) Soma size and basal dendritic branches of layer V

pyramidal neurons in Tsc2+/+ and Tsc2+/- mice. No differences were found in either soma size (lower left) and

the number of primary basal dendrites (lower right); (B) Effect of rapamycin on dendritic spine density in

Tsc1flox/flox and Tsc2+/+ control mice. Compared to DMSO vehicle controls, rapamycin treatment at these doses

between P21 and P30 did not show significant effect on spine pruning in either control mouse line. Data are

presented as mean± SD.

Figure S4. CamkII cre mediated recombination in mouse brain, related to Figure 3. (A) CamkII cre mice

were crossed to Rosa-YFP reporter mice. The distribution of cre mediated recombination was examined by

immunolabel for YFP at postnatal day P18, P20 and P23. No labeled cells were detected at deep cortical

layers at P18, and very few cells were observed in layer V cortex at p20. At P23, cre expression was fully

established in all cortical layers. (B) pS6 immunohistochemistry in Tsc1CKO mice at P18, P20 and P23.

Tsc1flox/flox mice were crossed to CamkII cre mice. The deletion of Tsc1 gene was determined by the level of

pS6, a downstream reporter for mTOR activity. In Tsc1flox/flox control mice, pS6 immunoreactivity peaked at P20

and decreased at P23. In Tsc1CKO mice, pS6 increased with age and was higher than in controls at P23,

consistent with the expression level of YFP cre reporter gene in (A); (C) western blot analysis of p-mTOR and

 
pS6 protein levels in Tsc1flox/flox controls and Tsc1CKO mice at postnatal days P21, P23 and P30; (D) Soma size

and basal dendritic branches in Tsc1flox/flox and Tsc1CKO mice. Compared to Tsc1flox/flox controls. Tsc1CKO mice

displayed a ~15% increase in soma size (E), * compared to Tsc1flox/flox, p<0.05, unpaired t test. No changes in

the number of primary basal dendrites were detected (F). Rapamycin slightly decreased soma size but not the

basal dendritic branches. Data are presented as mean± SD.

Figure S5. Impaired basal autophagy in Tsc deficient neurons related to Figure 4. (A) mTOR activity

and autophagy in Tsc2+/- mouse brain. Tsc2+/- mice were crossed to mice carrying GFP-LC3, a fluorescent

marker for autophagosomes. Cortical slices were immunolabeled for GFP and pS6, a downstream reporter for

mTOR activity. Tsc2+/- cortical pyramidal neurons displayed reduced GFP-LC3 fluorescence and GFP-LC3

puncta, indicating decreased autophagosomes in Tsc2+/- neurons; (B) LC3 and p62 immunolabel in primary

cortical neuronal cultures from wt and Tsc2+/- mice. Tsc2+/- neurons exhibited lower LC3 fluorescence and

more p62 puncta. Rapamycin normalized LC3 and p62 levels; (C-D) Accumulation of damaged mitochondria in

Tsc2+/- neurons. Primary cortical neuronal cultures from Atg7flox/flox; Nestin-Cre mice, in which Atg7 is depleted

in all CNS neurons, were used as controls for autophagy deficiency. Nestin promoter driven cre is expressed

embryonically, which ensures the depletion of Atg7 in these postnatally-derived cultured neurons. wt, Tsc2+/-,

Atg7flox/flox:Nestin-cre (Atg7CKO:Nestin-Cre) primary cortical neurons were stained with MitoTracker red (MitoRed)

and green (MitoGreen) (C). The uptake of MitoRed is dependent on mitochondrial membrane potential, while

MitoGreen shows the total mass of mitochondria. A significant decrease in the ratio of MitoRed (functional

mitochondria) to MitoGreen (total mass of mitochondria) in Tsc2+/- and Atg7CKO:Nestin-Cre neurons denoted an

accumulation of damaged mitochondria due to autophagy inhibition (D). * compared to wt, p<0.05; (E)

Accumulation of lipid droplets in Tsc2+/- and Atg7CKO:Nestin-Cre neurons. Compared to controls, Tsc2+/- and

Atg7CKO:Nestin-Cre neurons displayed higher Lipidtox Red intensity, demonstrating a higher content of lipid

droplets; (F) Autophagy level and mTOR activity in Tsc1CKO and control (Tsc1flox/flox) mouse brains. Tsc1flox/flox

and Tsc1CKO cortical sections were immunolabeled with antibodies against TSC1, pS6 and autophagy markers

LC3, p62 and Ub. Tsc1CKO mice showed loss of TSC1, increased mTOR activity as indicated by increased pS6,

 
and decreased basal autophagy as indicated by decreased LC3 fluorescence and accumulation of p62 and

Ub.

Figure S6. Behaviors, neuronal morphology, and glial markers in Atg7flox/flox and Atg7CKO mice, related

to Figure 5. (A-G) Open field measures of locomotor activity and anxiety-like behavior in Atg7CKO mice. (A)

Total distance traversed (meter, m); (B) time (second, s) spent immobile; (C) time (s) spent in the center arena;

(D) number of fecal boli; (E) time (s) spent self-grooming; (F) frequency of rearing during the test session; (G)

frequency of digging/foraging during the test session. No genotype and gender differences were found for any

parameter. n(control, male)=15; n(control, female)=20; n (Atg7CKO, male)=13; n(Atg7CKO, female) = 4. Data

are presented as mean ± SEM. (H-J) Soma size and basal dendritic branches in Atg7CKO mice. (H) A

representive DiI labeled layer V pyramidal neurons. No differences were observed for soma size (i) or number

of primary basal dendrites (J) between Atg7flox/flox and Atg7CKO mice. Data are presented as mean± SD. (K-L)

Markers for reactive astrocyte and microglia in Atg7CKO mice at different postnatal ages (P20, P30 and

9months (9mo)). (K) GFAP (Green), p62 (Red) and DAPI (Blue) immunolabel in Atg7CKO mice revealed no

significant increases in GFAP immunofluorescence; (L) microglial marker Iba 1 (Red) and DAPI (Blue)

immunolabel in Atg7CKO mice, showing no significant increases in Iba 1 in Atg7CKO brain. The positive control

indicates microglial activation in mouse hippocampus following pilocarpine induced seizures.

 
Supplemental Table S1 Demographic data for fixed brains used for Golgi study and immunohistochemistry
in Figures 1 & 2.

Sample Age Brain Brain PMI Length of Seizure


Group Gender Cause of Death
ID (Y) Weight PH (Hrs) Storage (D) History
A1 3 Male 1330 N/A 15 102.3 N N/A
ASD A2 7 Male 1560 N/A 11.4 84.0 N Drowning
(3-9yrs)
A3 7 Male 1610 N/A 25 77.0 Y Drowning
n=5
A4 7 Male N/A 5.9 3 47.4 N Complication of cancer

A5 9 Male 1320 N/A 27 127.0 Y Diffuse hemorrhage

C1 2 Female N/A 5.9 16 24.3 N Ingestion of drugs


Control C2 2 Male N/A 6.3 16 13.3 N Asphyxia, choking on food
(2-8yrs)
C3 5 Male N/A 6.27 17 120.0 N Drowning
n=5
C4 6 Female N/A 6.2 24 17.4 N Respiratory failure

C5 8 Male N/A N/A 16 13.7 N Blunt force neck injury

A6 13 Male 1470 N/A 8 111.0 Y Seizure suspected


ASD A7 14 Male 1615 N/A 10.3 158.0 N Heart stroke
(13-19yrs)
A8 15 Male 1090 N/A 26.8 74.0 Y Seizure
n=5
A9 15 Male N/A 6.3 9 45.9 N Drowning

A10 19 Male N/A N/A 22 46.8 Y N/A

C6 13 Male N/A 6.6 16 40.2 N Natural


Control C7 16 Male N/A 6.67 15 59.8 Y Multiple injuries
(13-18yrs)
C8 17 Male 1460 N/A 30.8 35.0 N Asphyxia
n=5
C9 17 Male 1250 N/A 28.9 N/A N N/A

C10 18 Male N/A N/A 21 96.9 N Cardiac Arrhythmia

No significant differences were detected in age, PMI and length of storage between patients and controls
from different age groups, p<0.05 for all unpaired t tests. A, ASD; C, control.

 
Supplemental Table S2 Correlations between spine density, western blot data and confounding factors
used in Figures 1 & 2.

Cause of Seizure Storage


Age PMI Gender
Death History Time

Spine -0.726 -0.235 0.087 -0.253 -0.057 0.197


r (p)
Density (0.000)* (0.318) (0.716) (0.283) (0.812) (0.420)

0.407 0.231 -0.218 -0.143 -0.054 0.210


p-mTOR r (p)
(0.006)* (0.132) (0.151) (0.348) (0.755) (0.232)

-0.419 -0.154 0.242 0.285 0.474 -0.243


LC3 II r (p)
(0.004)* (0.318) (0.109) (0.058) (0.004)* (0.166)

0.221 0.099 0.186 -0.226 -0.487 0.362


p62 r (p)
(0.249) (0.608) (0.335) (0.239) (0.007)* (0.065)
-0.433 -0.059 0.158 0.261 -0.036 0.163
PSD95 r (p)
(0.003)* (0.701) (0.300) (0.083) (0.835) (0.357)

r denotes spearman correlation coefficient; p in parentheses denotes significance p value.

 
Supplemental Table S3 Demographic data for postmortem frozen brain samples for western blot analysis
used in Figures 1 & 2.

Sample PMI Length of Seizure


Group Age Gender Cause of Death
ID (Hrs) Storage (D) History
A11 9 Male 27.0 127.0 Y Diffuse hemorrhage
A12 2 Male 4.0 77.0 N Drowning
ASD A13 5 Male 25.5 108.0 N Drowning
(2-9yrs) A14 5 Female 32.7 52.0 N Drowning
n=8 A15 8 Male 22.2 105.0 N Cancer
A16 4 Female 13.0 59.5 N Multiple injuries
A17 7 Male 3.0 26.0 N Cancer
A18 7 Male 20.0 47.9 N Drowning
C11 2 Female 16.0 24.3 N Asphyxia, Ingestion of drugs
C12 3 Female 12.0 83.7 N Drowning
Control
(2-9yrs) C13 3 Male 16.0 13.3 N Asphyxia, Choking on food
C14 5 Male 17.0 59.7 N Trauma
n=7
C15 5 Male 17.0 120.0 N Drowning
C16 5 Female 24.0 17.4 N Respiratory failure
C17 8 Male 16.0 13.7 N Blunt force neck injury
A19 20 Male 23.7 127.0 N Accident, multiple injuries
ASD
(13-20yrs) A20 16 Male N/A 82.0 Y Seizure
A21 19 Male 28.1 49.0 N Asphyxia, Choking on food
n=8
A22 14 Male 9.0 45.9 N Drowning
A23 16 Male 20.0 108.9 Y Diabetic Ketoacidosis
C18 13 Male 16.0 40.2 N Natural
C19 19 Male 9.0 106.7 N Multiple injuries
C20 19 Male 17.0 51.0 N Multiple injuries
Control
(13-20yrs) C21 19 Male 15.0 96.9 N Multiple injuries
C22 20 Male 12.0 90.4 N Multiple injuries
n=5
C23 16 Male 21.0 59.8 N Cardiac Arrhythmia
C24 17 Male 30.8 35.0 N Asphyxia
C25 19 Male 18.6 N/A N Pneumonia
C26 17 Male 28.9 N/A N N/A

No significant differences in age, PMI and length of storage were detected between patients and controls, p<0.05
for all unpaired t tests. The distribution of gender was matched between patients and controls aged 2-9yrs. A,
ASD; C, control.

 
Experimental Procedures

Postmortem human brains Fixed and frozen tissue from BA21 temoporal lobe in ASD and control subjects

were requested from Maryland Brain Bank, the Autism Tissue Portal, and Harvard Brain Bank bank, and were

based on tissue availability for specimens under the 21y. All patients met DSM-IV and ADI-R diagnosis for

autism. Demographic data for cases with frozen and fixed brain tissue is presented in Supplementary Tables 1

and 3. We examined only male adolescents to exclude the confounding effects of gender and levels of

hormone (Munoz-Cueto et al., 1990). Statistical differences in demographic parameters between patients and

controls were analyzed using an unpaired t-test.

Mouse strains Tsc2+/-, CamkII-Cre, Nestin-Cre mice were obtained from Jackson Labs and Atg7flox/flox and

GFP-LC3 mice were a gift of Maksaaki Komatsu (Tokyo Metropolitan Institute of Medical Sciences), all on the

C57Bl/6J clonal background. We crossed Atg7flox/flox mice with CamkII Cre mice to obtain mice specifically

deficient in autophagy in excitatory neurons (Atg7CKO) or with Nestin-Cre lines to obtain pan-neuronal

autophagy deficient mice (Atg7CKO:Nestin-Cre). Tsc2+/- mice were bred into GFP-LC3 mice for measuring

autophagic activity. Tsc2+/- mice were bred into Atg7flox/+ :CamkII Cre+ mice to generate Tsc2 mutant;

autophagy deficient mice (Tsc2+/-: Atg7CKO). Forebrain-specific Tsc1-deficient mutant (Tsc1CKO) mice were of

mixed genetic backgrounds (C57Bl/6J, 129 SvJae), generated by crossing Tsc1flox/flox mice (129 SvJae) with

CamkII Cre mice (C57Bl/6J). All mouse experimental procedures were reviewed and approved by Columbia

University Medical Center Institutional Animal Care and Use Committee.

Behavioral tests All mice were tested in open-field for anxiety-like/motor behavior, assessed during dyadic

social interactions, and tested in the 3-chamber apparatus for social investigation (Bolivar et al., 2007;

Winslow, 2003). Self-grooming behavior was also assessed. All tests were conducted under white light

between 9.00 and 17.00.

Open Field Levels of anxiety and general exploratory locomotion in a novel open field environment was

assessed, as an independent control for changes of physical activities that could confound the interpretation of

results from the self-grooming and social approach tasks. Measures were: total distance traveled (m), total

time immobile (s), time in inner area (s), Rearing and number of fecal boli produced. Males and females were

 
tested on separate days to avoid odor preferences. Videos were later analyzed with the automated tracking

program ANY-Maze (v4.82, Stoelting).

Novel Object Recognition Subjects were habituated for ten minutes in one of four identical, translucent blue

arenas (18.25”x 13.87”x 10.87”). Each apparatus was cleaned with a 30% warm soap solution and contained

fresh bedding for each trial. One day after habituation, the mouse was reintroduced to the chamber, now

containing two identical objects (yellow rubber ducks) placed in opposite corners of the apparatus. Tests were

video recorded for 15 minutes, after which the mouse was returned to the home cage. Thirty minutes after the

initial test, the mouse was placed into the arena, now containing one original object and one novel object

(yellow cylinder), and behavior was recorded for 15 minutes. Video recorded sessions were coded for time

spent actively sniffing the novel object vs. the original/familiar object.

Dyadic social Interaction Male mice were assessed for dyadic social interactions with unfamiliar (non-

cagemates) mice matched for sex and genotype. One day prior to testing, tails were marked with a non-toxic

marker for identification during analysis. Each animal was placed in an 18.25”x 13.87”x 10.87” arena. Subjects

were brought into the testing room individually, in clean transfer cages, and were placed in opposing corners of

the chamber simultaneously. Behavior was video recorded for 15 minutes and was coded by two researchers

blind to genotype. Minute-by-minute analysis of the number of observed bouts of sniffing, walking, rearing, tail

rattling, dig/foraging, huddling, grooming, biting, mounting as well as total time spent sniffing were measured.

Three Chamber tests for sociability and preference for social novelty Mice were tested in a three-

chambered arena for three 10-min phases: (1) Habituation: the test mouse was first placed in the middle

chamber and allowed to explore, with the doorways into the two side chambers open. (2) Sociability: following

the habituation period, the test mouse was enclosed in the center compartment of the social test box, and an

unfamiliar mouse was enclosed in a wire pencil cup placed in a side chamber. The location for the unfamiliar

mouse alternated between the left and right sides of the social test box across subjects. An empty wire pencil

cup was placed in the opposite side, to serve as a novel object control. Following placement of the mouse and

cup, the doors were re-opened, and the subject was allowed to explore the entire social test box. Measures

were taken of the amount of time spent sniffing the mouse and the empty cup. (3) Preference for social

novelty: at the end of the sociability test, each mouse was further tested for preference to spend time with a
 

 
novel mouse. A new unfamiliar mouse (novel mouse) was placed in the wire pencil cup that had been empty

during the previous session. The test mouse then had a choice between the first, already-investigated mouse

(familiar mouse) and the novel unfamiliar mouse (novel mouse). The same measures were acquired as for the

sociability test.

Self-grooming Mice were scored for spontaneous grooming behaviors. A thin (1 cm) layer of bedding

reduced neophobia, while preventing digging; a potentially competing behavior. After a 10-min habituation

period in the test cage, each mouse was scored with a stopwatch for 10 min for cumulative time spent

grooming all body regions.

Immunohistochemistry 10 controls and 10 ASD patients were included for immunohistochemistry on 7µm

thick paraffin-embedded temporal lobe brain sections. Triplicate cryosections (7µm thick) with comparable

anatomy were chosen from mutants and corresponding control mice. Antibodies used for

immunocytochemistry were against LC3 (1:200, Novus), p62 (1:300, American Research Products), ubiquitin

(Ub, 1:300, Millipore), PSD95 (1:100, Abcam) and synaptophysin (1:300, Abcam).

Neuronal culture and immunocytochemistry Cortical neurons from postnatal day1-3 mice were

dissociated and plated on primary rat astrocyte monolayers. At day 14-15 in vitro, primary neurons were fixed

with 4% paraformaldehyde or subjected to live cell staining for lipid droplets (LipdtoxRed, Invitrogen) and

mitochondria (MitoTracker Red and Green, Invitrogen). Fixed neuronal cultures were immunostained with

antibodies against autophagy markers LC3 (1:200, Novus), p62 (1:500, American Research Products), PSD95

(1:100, Abcam) and F-actin (1:50, Alexa Fluor 488 phalloidin, Invitrogen). For spine dynamic analysis,

hippocampal neurons were cultured at low density. At day 14-15 in vitro, neurons were infected with an EGFP

expressing control virus or Atg7SiRNA-EGFP virus. Neurons were fixed for Immunocytochemistry at DIV 19-20

or were live imaged for spine formation or elimination during a 12 hour time window.

Western blotting Mouse and human brain tissue were lysed with 1X RIPA buffer supplemented with

protease inhibitors and phosphatase inhibitors. Homogenates were centrifuged at 14000rpm at 4C for 30min

and supernatant were collected for western blotting. Antibodies used were against LC3 (1:1000, Novus), p-

mTOR(1:1000, Cellsignal), t-mTOR(1:1000, Cellsignal), PSD95(1:2000, Abcam), p-S6(1:1000, Cellsignal), t-

 
S6(1:1000, Cellsignal), Atg12 & Ag5 (1:1000, Cellsignal), p62(1:2000, American Research Products), and actin

(1:3000, Sigma).

Golgi impregnation and image analysis Neuronal morphology in postmortem human brain was analyzed

using Golgi-Kopsch technique, as described previously (Rosoklija et al., 2003). Thirty-two subjects, ranging in

age from 2y to 20y were included with 20 samples successfully stained (success rate ~62%). In all cases, only

neurons in which the basal dendritic tree was completely stained were included in this analysis. 3-D

reconstruction ensures spines located on the top or bottom surface of the dendrites could be analyzed,

avoiding underestimate of the number of spines and of the spine densities. Because spine density and possibly

spine size changes as a function of distance from the soma (Benavides-Piccione et al., 2002), we compared

similar segments of dendrites between different cells by selecting segments of basal dendrites which were

located at the same proportional distance from the soma. We selected basal dendrites segments that were 70-

100 µm distant from the soma, immediately after the first branching point. 15-30 isolated layer V pyramidal

cells per tissue block were randomly chosen for analysis and 3-4 dendritic segments per cell were analyzed.

Images were acquired using an Olympus BX21 system equipped with a Z axis control and a 100X oil long

working distance objective. The acquired brightfield images were pre-processed with ImageJ to eliminate

background pixels. The resulting transparent “false fluorescent” Z-stacks were reconstructed using Imaris

software FilamentTracer Module (Bitplane). Spine density was represented by average number of spines per

10 µm of dendritic length. Spine density, length of spine neck and spine head diameter was analyzed using

ImarisXT module (Bitplane).

DiOlistic labeling of dendritic spine in mouse brain and imaging Anesthetized mice were transcardially

perfused with 4% PFA. Whole brains were removed and sectioned at 200 µm thickness on a vibratome. Brain

slices were labeled with DiI using a Helios gene gun system at 120 psi. Z stack images with 0.2 µm step size

were acquired with a Leica multiphoton system. The basal dendritic segments were imaged using the same

criteria described above. Fluorescent Z-stacks were reconstructed using Imaris software FilamentTracer

module (Bitplane) and spine analysis was performed using ImarisXT module. Primary auditory cortex (A1) and

secondary somatosensory cortex (S2), approximately the temporal cortices of mice, were selected as reported

previously (Benavides-Piccione et al., 2002).


 

 
Statistical analysis Statistical analyses were performed using Graphpad Prism software using one-way or

two-way ANOVA with Bonferroni’s multiple comparison tests for post-hoc analysis. Normal distribution of data

was determined using the Kolmogorov-Smirnov test. Differences between two groups were analyzed by two-

tailed t-test. Correlations were analyzed using Spearman correlation test and nonlinear regression. Results are

presented as mean ± SEM or mean ± S.D. p < 0.05 was considered statistically significant.

 
References for Experimental Procedures

Winslow, J. T. (2003). Mouse social recognition and preference. in Curr. Protoc. Neurosci., Crawley, J.N., et
al., ed. (John Wiley & Sons. Inc, New york), pp. 8.16.1-8.16.16.
Bolivar, V. J., Walters, S. R. & Phoenix, J. L. (2007). Assessing autism-like behavior in mice: variations in
social interactions among inbred strains. Behav. Brain Res. 176: 21-26.
Munoz-Cueto, J. A., Garcia-Segura, L. M. & Ruiz-Marcos, A. (1990).Developmental sex differences and effect
ofovariectomy on the number of cortical pyramidal cell dendritic spines. Brain Res. 515, 64-68.
Benavides-Piccione, R., Ballesteros-Yáñez, I., DeFelipe, J., Yuste, R. (2002). Cortical area and species
differences in dendritic spine morphology. J Neurocytol. 31:337-346.
Rosoklija, G., Mancevski, B., Ilievski, B., Perera, T., Lisanby, S.H., Coplan, J.D., Duma, A., Serafimova, T.,
Dwork, A.J. (2003). Optimization of Golgi methods for impregnation of brain tissue from humans and
monkeys. J. Neurosci. Methods 131, 1-7.

You might also like