differential mitochondrial requirements for radially and ... · these two neuronal cell populations...

24
Report Differential Mitochondrial Requirements for Radially and Non-radially Migrating Cortical Neurons: Implications for Mitochondrial Disorders Graphical Abstract Highlights d Mitochondria in cortical interneurons are motile during migration d Mitochondria are stationary in migrating cortical projection neurons d Oxidative phosphorylation defects disrupt cortical interneuron migration d Interneurons lose polarity when oxidative phosphorylation is perturbed Authors Erika G. Lin-Hendel, Meagan J. McManus, Douglas C. Wallace, Stewart A. Anderson, Jeffrey A. Golden Correspondence [email protected] (S.A.A.), [email protected] (J.A.G.) In Brief Mitochondrial disorders frequently result in neurological dysfunction, but the causes of pathogenesis are uncertain. Lin-Hendel et al. show that mitochondrial dysfunction during neurodevelopment selectively disrupts cortical interneuron migration and not projection neuron migration. They further show a specific dependence on oxidative phosphorylation of interneuron migration. Lin-Hendel et al., 2016, Cell Reports 15, 229–237 April 12, 2016 ª2016 The Authors http://dx.doi.org/10.1016/j.celrep.2016.03.024

Upload: others

Post on 08-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Report

Differential Mitochondrial

Requirements for Radiallyand Non-radially Migrating Cortical Neurons:Implications for Mitochondrial Disorders

Graphical Abstract

Highlights

d Mitochondria in cortical interneurons are motile during

migration

d Mitochondria are stationary in migrating cortical projection

neurons

d Oxidative phosphorylation defects disrupt cortical

interneuron migration

d Interneurons lose polarity when oxidative phosphorylation is

perturbed

Lin-Hendel et al., 2016, Cell Reports 15, 229–237April 12, 2016 ª2016 The Authorshttp://dx.doi.org/10.1016/j.celrep.2016.03.024

Authors

ErikaG. Lin-Hendel,Meagan J.McManus,

Douglas C.Wallace, Stewart A. Anderson,

Jeffrey A. Golden

[email protected] (S.A.A.),[email protected] (J.A.G.)

In Brief

Mitochondrial disorders frequently result

in neurological dysfunction, but the

causes of pathogenesis are uncertain.

Lin-Hendel et al. show that mitochondrial

dysfunction during neurodevelopment

selectively disrupts cortical interneuron

migration and not projection neuron

migration. They further show a specific

dependence on oxidative

phosphorylation of interneuronmigration.

Page 2: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Cell Reports

Report

Differential Mitochondrial Requirementsfor Radially and Non-radially Migrating CorticalNeurons: Implications for Mitochondrial DisordersErika G. Lin-Hendel,1,2 Meagan J. McManus,3,4 Douglas C. Wallace,3,4 Stewart A. Anderson,5,7,* and Jeffrey A. Golden6,7,*1School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA2Developmental, Regenerative and Stem Cell Biology, Biomedical Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA3Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA4Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA5Department of Psychiatry, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA6Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA7Co-senior author

*Correspondence: [email protected] (S.A.A.), [email protected] (J.A.G.)http://dx.doi.org/10.1016/j.celrep.2016.03.024

SUMMARY

Mitochondrial dysfunction has been increasinglylinked to neurodevelopmental disorders such as intel-lectual disability, childhood epilepsy, and autismspectrum disorder, conditions also associated withcorticalGABAergic interneurondysfunction. Althoughinterneurons have some of the highest metabolicdemands in the postnatal brain, the importance ofmitochondria during interneuron development isunknown. We find that interneuron migration fromthe basal forebrain to the neocortex is highly sensi-tive to perturbations in oxidative phosphorylation.Both pharmacologic and genetic inhibition of adeninenucleotide transferase 1 (Ant1) disrupts the non-radialmigration of interneurons, but not the radial migrationof cortical projection neurons. The selective depen-dence of cortical interneuron migration on oxidativephosphorylation may be a mechanistic pathwayupon which multiple developmental and metabolicpathologies converge.

INTRODUCTION

Mitochondrial diseases (MDs) are the most common inherited

metabolic disorder, with an estimated prevalence of 1:5,000

(Schaefer et al., 2004). Although MDs consist of a spectrum of

disorders that can involve single or multisystem presentations,

neurological symptoms are common clinical characteristics. In

recent years, clinical, genetic, and biochemical studies have re-

vealed an emerging link between mitochondrial dysfunction and

neurodevelopmental disorders, including intellectual disability

(ID) (Valenti et al., 2014), childhood epilepsy (Chevallier et al.,

2014), and autism spectrum disorder (ASD) (Rossignol and

Frye, 2012). Interestingly, these conditions have also been asso-

ciated with interneuron dysfunction (Marın, 2012). The correla-

tion between MDs and childhood neurological disorders raises

This is an open access article under the CC BY-N

the question as to whether interneuron development is particu-

larly dependent on mitochondrial function.

Recent studies have elucidated roles for mitochondria in mul-

tiple aspects of neurodevelopment including neuronal differenti-

ation (Wang et al., 2014), process outgrowth (Cheng et al., 2012;

Kimura and Murakami, 2014), and synaptogenesis (Bertholet

et al., 2013). Most of these studies have utilized glutamatergic

hippocampal neurons as a model, leaving the contribution of

mitochondria to interneuron development relatively unexplored.

Since interneurons are thought to be a key factor in the patho-

genesis of epilepsy and ASD, we were particularly interested in

examining mitochondrial behavior and function during develop-

mental processes for which there are distinct features between

glutamatergic projection neurons (PNs) and GABAergic inter-

neurons (INs) in the cortex. During development, PNs and INs

are derived from distinct locations and take different migration

routes: PNs migrate along radial glial fibers from their origin in

the dorsal ventricular zone and maintain a relatively stable

morphology oriented toward the pial surface (Noctor et al.,

2004). In contrast, cortical INs take a circuitous path from their

origin in the subcortical ganglionic eminences. Along the way,

migrating INs frequently pause, change direction, and exhibit

extensive branching dynamics of the leading process (Bellion

et al., 2005; Lysko et al., 2011, 2014; Polleux et al., 2002).

Given the greater distance and dynamic nature of IN migra-

tion, we reasoned that it requires more energy than does

the migration of PNs. First, we found that the mitochondria within

INs have a highly dynamic localization pattern during non-radial

migration. In contrast, mitochondria remain more consistently

localized in radially migrating PNs. Second, we found migrating

interneurons to be exquisitely sensitive to agents that block

the utilization of ATP generated through oxidative phosphoryla-

tion. Remarkably, glycolysis alone is insufficient for normal

IN migration but is able to support the radial migration of PNs.

Moreover, the genetic disruption ofmitochondrial oxidative phos-

phorylation (OXPHOS) in mice lacking Ant1 was associated with

dramatic alterations of IN migratory morphology and behavior,

includingmispositioningof thecentrosomes.Conversely,Ant1�/�

PNs appeared normal in our migration assays. These data

Cell Reports 15, 229–237, April 12, 2016 ª2016 The Authors 229C-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Page 3: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Figure 1. Mitochondrial Localization in Migrating Neurons

(A) Schemata of interneuron (IN; 1–3) morphologies displayed during migration.

(B) Confocal immunofluorescence (IF) images of mitochondria in migrating INs in vitro displaying varying localization patterns according to morphology.

Cytosol (GFP), mitochondria (Tom20), and nuclei (DAPI). Scale bar, 10 mm.

(C) Quantification of clustering of mitochondria in subcellular locations. Region I, trailing process (TP); II, overlapping nucleus (Nuc); III, 5 mm anterior to nucleus

(5 mm AN); IV, cytoplasmic bleb (bleb); V, leading process (LP); and VI, leading process tip (LPT). Clustering varied markedly between IN morphologies. IN

morphology 1 clustered in III (*p = 0.039). IN morphology 2 clustered in IV (*p = 0.02), while IN morphology 3 clustered in I and II (*p = 0.0343 and ***p = 0.0002).

Error bars represent median with 25th–75th percentiles ± min/max value of percent total mitochondrial area (%TMA) normalized to region’s percent of total cell

area (%TCA). n = 15 cells each type; Freidman’s test with Dunn’s correction.

(D) Schemata of pyramidal neuron (PN) migration morphology.

(legend continued on next page)

230 Cell Reports 15, 229–237, April 12, 2016

Page 4: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

suggest that interneuron polarity during migration is particularly

sensitive to disruptions in metabolism, and that OXPHOS is

required for normal migration of INs but not PNs. Our results

also imply that the symptomatic manifestations of mitochondrial

dysfunction and related conditions, including hypoxic injury, on

cerebral cortical function may be secondary to their selective

impact on cortical interneuron migration.

RESULTS

Mitochondria Are Highly Dynamic during InterneuronMigrationTo examine the role of mitochondria in non-radial versus radial

migration, we first sought to characterize the localization of

mitochondria in migrating INs and PNs. We classified medial

ganglionic eminence (MGE)-derived cells migrating in explant

cultures into three morphological classes corresponding to

distinct phases of their migration: leading process extension, for-

ward movement of the centrosome, and nucleokinesis/trailing

process retraction (Marın et al., 2006). Morphology 1 cells were

defined to have slender, tapered leading processes; morphology

2 cells have a bleb or thickening of the leading process; and

morphology 3 cells have a clear trailing process (Figure 1A).

Each subgroup displayed a distinct distribution of mitochondria

(Figure 1B). In morphology 1, mitochondria were concentrated

immediately anterior to the nucleus (Figure 1C; 49 ± 4, % total

mitochondrial area [TMA] ± SEM, 5-fold greater % TMA% total

cell area [TCA], p < 0.05) (Bellion et al., 2005; Golden et al.,

1997). In morphology 2 cells, mitochondria were concentrated

in the cytoplasmic bleb (Figure 1C; 71 ± 2, %TMA ± SEM,

3.9-fold greater %TMA/%TCA than other areas, p < 0.05), while

in morphology 3, the mitochondria were aggregated in the trail-

ing process and posterior nuclear area (Figure 1C, 29 ± 4, %

TMA ± SEM, 39% ± 5 TMA ± SEM respectively, 1.7-fold greater

%TMA/%TCA than other areas, p < 0.05) (Bellion et al., 2005).

Although these independent clustering behaviors have been

noted in the literature, mitochondrial dynamics during migration

have not been studied. To evaluate the subcellular localization of

mitochondria in relation to the morphological migratory phases,

we next performed time-lapse imaging of fluorescently labeled

mitochondria in migrating interneurons. Interestingly, mitochon-

dria displayed consistent positional reorganization during migra-

tion, as their subcellular location changed in concert with the

morphology of the migrating cell (Figures 1G and 1H; Movie

S1). The mitochondrial localization and changes in location

observed in the three IN morphologies were confirmed by live

(E) Confocal IF images of mitochondria in representative migrating PN. Cytosol (

(F) Quantification of clustering mitochondria in migrating PNs. Region I: TP; II: Nu

*p = 0.0327, **p = 0.0064, and ****p < 0.0001. n = 15 cells; Freidman’s test with

(G) Time-lapse imaging of a migrating Dlx5/6CIG IN in vitro (cytosol, GFP; mitochon

1 frame = 10 min. Scale bar, 10 mm. See also Movie S2.

(H) Quantification of INs displaying a extensive movement of mitochondrial th

compartment. The majority of INs showed this movement of mitochondria throu

cultures, 200 cells. Values represent mean ± SEM.

(I) Live time-lapse imaging of a migrating PN (cytosol, GFP; mitochondria, MitoD

(J) Quantification of PNs movement of mitochondrial through the cell versus tho

movement of mitochondria through PNs was essentially not observed. p = 0.

mean ± SEM.

imaging of mitochondria in whole-brain-slice cultures from em-

bryonic day 13.5 (E13.5) embryos (Figure S1; Movie S2). The

localization of mitochondria in migrating INs was ranked accord-

ing to morphology as matching or not matching the localization

observed in migrating dissociated INs and found to be highly

correlated (Figure S1A).

In contrast tomigrating INs,migrating PNsmaintain a relatively

consistent migratory morphology after leaving their multipolar

phase in the ventricular and subventricular zones (Figure 1D

and 1I) (Noctor et al., 2004). During radial migration in the cortical

plate, mitochondria were found primarily anterior to the nucleus

and in the leadingprocess, showing little change in regional local-

ization (Figures 1E, 1F, 1I, and 1J; Movie S3). These data reveal

that the intracellular position, and changes in location, of mito-

chondrial of INs and PNs are clearly distinguishable and suggest

that there may be differences in energy requirements between

these two neuronal cell populations during development.

Oxidative Phosphorylation Is Necessary for Normal INMigration, but Not for Radial MigrationTo determine whether migrating INs and PNs have distinct ener-

getic requirements, we studied their need for mitochondrially

generated ATP in explant and slice cultures. Cells generate

ATP through glycolysis in the cytosol and oxidative phosphoryla-

tion (OXPHOS) in the mitochondria. To test whether OXPHOS is

necessary for normal neuronal migration, we examined cell

movement after blocking OXPHOS with either oligomycin or

bongkrekic acid (BA). Oligomycin (Olig) blocks mitochondrial

production of ATP by inhibiting the ATP synthase (Kulka and

Cooper, 1962), while BA prevents the translocation of ATP

across the inner mitochondrial membrane by inhibiting the

adenine nucleotide translocator isoforms 1 (Ant1, also known

as Slc25a4) and 2 (Ant2 also known as Slc25a5) (Henderson

and Lardy, 1970). IN migration was exquisitely sensitive to Olig

treatment, where 0.02 mM reduced IN migration by 78% (Fig-

ure S2A; Movie S4). Treating INs with 2.5 mM BA reduced IN

migration by 50% (p% 0.001) (Figure 2A; Movie S5). These cells

showed no reduction in somal translocation (Figure 2C; not sta-

tistically significant but trending toward slower) but a significant

increase in the time spent paused (Figure 2D). Interestingly,

treated cells exhibited elongated trailing processes (Figures 2B

and 2F) as well as a higher frequency of trailing processes (Fig-

ure 2E) and for more time (Figure 2G) than controls. Furthermore,

at low BA concentrations, migrating INs exhibit a 10-fold in-

crease in direction changes (Figure 2H). BA also resulted in sig-

nificant reduction in the leading process length (Figure 2I). The

GFP), mitochondria (MitoDsRed), and nuclei (DAPI). Scale bar, 10 mm.

c; III: 5 mm AN; IV: cytoplasmic bleb (bleb); V: LP; VI: leading process tip LPT.

Dunn’s correction.

dria, MitoTracker Red CMXRos) show intracellular movement of mitochondria.

rough the cell versus those where the mitochondria remain confined to one

ghout the cell during migration. p < 0.0001, unpaired t test, n = 5 independent

sRed). 1 frame = 10 min. Scale bar, 10 mm. See also Movie S3.

se where the mitochondria remain confined to one compartment. Intracellular

002, Mann-Whitney, n = 6 independent cultures, 82 cells. Values represent

Cell Reports 15, 229–237, April 12, 2016 231

Page 5: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Figure 2. Pharmacological Inhibition of

OXPHOS Reduces IN Migration

(A) MGE explants were cultured in high

glucose ± BA, an inhibitor of OXPHOS. Inhibiting

OXPHOS decreased IN migration. (BA con-

centration: 0 mM and 0.5 mM = 0.36 ± 0.4;

2.5 mM = 0.18 ± 0.02, 5 mM = 0.09 ± 0.01; all in

mm/min ± SEM; 0 versus 2.5 or 5, p % 0.001,

n = 5 independent cultures, >150 cells each,

ANOVA test).

(B) Representative phase image of BA-treated

MGE INs. Arrows identify elongated trailing pro-

cesses. Scale bar, 50 mm.

(C) Somal translocation was calculated for

each migrating IN; while slightly slower, this

did not reach significance (BA concentration:

0 mM = 0.53 ± 0.04; 2.5 mM = 0.45 ± 0.01; all in

mm/min ± SEM; p % 0.075, n = 5 independent

cultures, > 150 cells each, unpaired t test,

Welch’s correction).

(D) The time a migrating neuron was paused

versus moving was also calculated. In the pres-

ence of BA, the neurons spend significantly

more time paused compared to control cells

(BA concentration: 0 mM = 0.30 ± 0.05; 2.5 mM =

0.59 ± 0.05; all in total time paused/imaging

period ± SEM; p % 0.002, n = 5 independent

cultures, > 150 cells each, unpaired t test, Welch’s

correction).

(E–G) In addition to decreased migration, inhibi-

tion of OXPHOS increased the frequency (E; p <

0.001, unpaired t test with Welch’s correction,

mean ± SEM), length (F; p < 0.0001, Mann Whit-

ney, median with interquartile range ± min/max),

and life of trailing processes (G; p < 0.0001, Mann-

Whitney). For each, n = 5 independent cultures,

100 cells each.

(H) OXPHOS inhibition also causes increases in

direction changes (the mean number of direction

changes/hour ± SEM was 0.024 ± 0.006 for 0 mM

BA; and 0.23 ± 0.05 for 2.5 mM BA, p < 0.0001;

n = 5 independent cultures, 100 cells each, un-

paired t test with Welch’s correction).

(I–K) Leading process length (I), branches/cell

(J), and branch length (K) were all signifi-

cantly reduced in migrating INs treated with

BA when compared to controls (p < 0.0001, p <

0.001, and p < 0.0001, respectively, Mann-Whit-

ney test).

(L) Sample migration paths of INs (yellow) and PNs

(red) in E16 brain slices treated with vehicle or 20 mM BA. See also Movie S5. CP, cortical plate; IZ, intermediate zone; VZ, ventricular zone. Scale bar, 150 mm.

(M) IN migration rates decreased in slices treated with BA (p = 0.0023, n = 6 individuals, 20 INs each) whereas PNs were unaffected (p > 0.05, n = 6 individuals,R

15 PNs in each). Unpaired t test with Welch’s correction, mean ± SEM.

leading processes also branched less frequently and the branch

lengths were significantly reduced (Figures 2J and 2K).

To determine whether OXPHOS is sufficient to supply energy

for IN migration, we removed glucose (GLUC) from the medium

or inhibited glycolysis with 2-deoxyglucose (2-DG) and pro-

vided the OXPHOS substrate pyruvate (PYR). Alternatively,

we substituted GLUC for galactose (GAL). GLUC deprivation

reduced migration by �53% (Figure S2C), while inhibition with

2-DG reduced migration by �68% (Figure S2C). When INs are

supplemented with PYR or GAL, ATP generation is dependent

on OXPHOS alone (Adeva-Andany et al., 2014; Marroquin

232 Cell Reports 15, 229–237, April 12, 2016

et al., 2007). Both PYR and GAL were sufficient to fully rescue

IN migration under conditions of glycolysis inhibition, and this

ability to sustain migration was abrogated with addition of sub-

threshold doses of BA (Figure S2C). Therefore, OXPHOS is

both necessary and sufficient for normal IN migration.

The requirement for OXPHOS in neuronal migration was

further tested using slice cultures where both non-radially

migrating INs, and radially migrating PNs, could be studied

simultaneously. We utilized Dlx5/6CIG mice in which the INs are

genetically labeled with EGFP. At E14, a DsRed expression

construct was electroporated into Dlx5/6CIG embryos to label

Page 6: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Figure 3. Interneuron Migration Is Reduced

in Embryonic Ant1 Mutants

(A) At E13.5 migrating INs (labeled by calbindin

immunohistochemistry) have not traveled as far in

Ant1�/� brains compared to Ant+/+ brains. The

white arrow indicates the leading migrating INs.

Ant1+/+servedas thestandardizedcontroldistance

of 1.0 ± 0.02 with Ant1�/� IN migration showing on

average 0.85 ± 0.02 relative distance units ± SEM

or a 15% reduction, p < 0.0001. Scale bar, 200 mm.

(B)Quantificationof relativedistanceof leading cells

ofmigrating INs. ****p < 0.0001, n = 5 individuals, 25

cells each, Mann-Whitney test. Values represent

median with interquartile range ±min/max.

(C) Quantification of relative INs in cortex ;

Ant1+/+ = 1.0 ± 0.06; Ant1�/� = 0.7 ± 0.04;

normalized INs in cortex ± SEM, **p = 0.004, n = 5,

unpaired t test with Welch’s correction.

(D) Ant1�/� INs in the cortex have increased

trailing process (TP) length. Ant1+/+: 1.1 mm ± 0.3;

Ant1�/�: 5.2 mm ± 1.1 (±SEM), *p = 0.022, n = 5

individuals, 25 cells each, unpaired t test with

Welch’s correction.

(E) Proliferation in germinal ventricular zone (VZ),

indicated by Ki67 immunostaining, was not

impacted by loss of Ant1. Medial ganglionic

eminence (MGE) and pallium (Ctx). Ant1+/+ versus

Ant1�/� for each region, p = 0.286 for MGE, p =

0.309 for Ctx; n = 5 individuals, unpaired t test with

Welch’s correction.

(F) Loss of Ant1 did not increase cell death, indi-

cated by caspase-3, in the MGE and the ventral

and dorsal pallium (V/D Ctx). Ant1+/+ versus

Ant1�/� for each region, p > 0.99, n = 5 in-

dividuals, Kruskal-Wallis, Dunn’s correction.

Values represent median, 25th–75th percentile ±

min/max.

(G) INs in cortex of Ant1�/� mice displayed abnormal leading process orientation. Arrowheads indicate misaligned INs. Scale bar, 75mm.

(H) Quantification of cortical IN leading process orientation into quadrants: Q1, dorsal; Q2, pial; Q3, ventral; and Q4, ventricular orientation. The average percent

of IN oriented in Q1 for Ant1+/+ was 80 ± 4 and 50 ± 4; Ant1�/� (±SEM), ****p = 0.0001 and **p = 0.0069, n = 5, ANOVA with Sidak’s correction.

the progenitors of radially migrating PNs. Embryos were har-

vested at E16, and cortical slices from individuals were treated

with BA. Remarkably, BA treatment reduced IN migration rates,

whereas PNs were unaffected (Figures 2L and 2M; Movie S6;

IN: 0.289 ± 0.06 mm/min decrease, p < 0.002; PN: 0.03 ±

0.02 mm/min increase, p > 0.1). These data demonstrate that

the non-radial migration of cortical interneurons is dependent

onOXPHOS, while the radial migration of cortical projection neu-

rons is either not or minimally OXPHOS dependent.

Selective Disruption of Non-radial IN Migration in Ant

1�/� MutantsWenext sought to corroborate our pharmacologic data of the dif-

ferential effects of OXPHOS on PN versus IN migration in mice

lacking the Ant1 isoform. The genetic removal of Ant1 reduces

the ATP flux from themitochondria to the cytosol, andAnt1 is ex-

pressed in cortical neurons, including INs (Figures S3A and S3B;

Graham et al., 1997; Lee et al., 2009; Levy et al., 2000).

To determine if loss of Ant1 disrupts IN migration in vivo,

Ant1+/+ and Ant1�/� brains were sectioned and stained for cal-

bindin at E13.5 to detect a subset of migrating INs. On average,

the leading edge of INs from wild-type brains migrated 15%

percent farther than that of Ant1�/� brains (Figures 3A and 3B),

and there was a 30% decrease in total migrating INs invading

the cortex in Ant1�/� animals (Figure 3C). Additionally, Ant1�/�

migrating INs displayed aberrant orientation of the leading pro-

cess with 30% fewer cells oriented in the main migration path

(Figures 3G and 3H). Interestingly, Ant1�/� INs had longer trailing

processes in vivo (Figure 3D), a characteristic also seen in wild-

type INs treated with BA in vitro (Figure 2C). Neither proliferation

nor cell death in the MGE or cortex was impacted by loss of

Ant1�/� (Figures 3E and 3F), excluding these mechanisms as

an explanation for the reduction of cells in the mutant cortex.

This suggests that a migration phenotype is the primary cause

for the IN defect in Ant1�/� embryonic cortex.

Toconfirm theeffectofAnt1deficiencyon INmigration,Ant1�/�

MGE explants were assayed. Ant1�/� INs migrated shorter dis-

tances (�40% decrease; Figures 4A and 4B), and more slowly

(�42%decrease;Figure 4C)whencompared toAnt1+/+ INs. Inter-

estingly, Ant1�/� IN migration was exquisitely sensitive to BA

treatment, showing large reductions in INmigration rates at doses

that hadnoeffect onAnt1+/+ cells (Figures 4C), likely due to further

blocking ofmitochondrial ATP efflux through inhibition of theAnt2

isoform (Graham et al., 1997; Levy et al., 2000).

Cell Reports 15, 229–237, April 12, 2016 233

Page 7: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Figure 4. Abnormal Migration by Ant1�/�

Interneurons

(A) MGE explants were cultured in vitro for 16 hr.

Ant1�/� INs did not migrate out of the explant as

far as controls.

(B) Quantification of relative distance of IN migra-

tion from explant showing a statistically significant

difference in the distance migrated between

Ant1�/� and Ant1�/� INs., Ant1+/+ is calculated as

the normalized distance (1.0 ± 0.03) and Ant1�/�

was on average approximately 40% reduced

(0.61 ± 0.08); normalized distance ± SEM. **p =

0.0034, n = 6, 50 cells each genotype, unpaired t

test with Welch’s correction. Scale bar, 250 mm.

(C) Quantification of INmigration rates of wild-type

and mutant INs. Ant1�/� IN migration rates had

slower migration rates (****p < 0.0001) and were

more sensitive to BA treatment compared to

Ant1+/+(****p < 0.001). Migration rates Ant1+/+:

0.42 ± 0.01; Ant1�/�: 0.24 ± 0.01; mm ± SEM, n =

5, 150 cells each condition, ANOVA with Bonfer-

onni’s correction.

(D) Examples of migration path of GFP+ INs in slice

culture (also see Movie S7). Dots, start; lines,

paths. Scale bars, 150 mm.

(E) Ant1�/� INs in slices display decreased

migration rates relative to control. The relative

migration rate was compared between Ant1+/+:

0.94 ± 0.04 and Ant1�/�: 0.65 ± 0.04; relative

migration rate ± SEM, ****p < 0.0001, n = 5, un-

paired t test with Welch’s correction.

(F) Ant1�/� INs display increased direction

changes. Ant1+/+: 0.07 ± 0.01; Ant1�/�: 0.25 ±

0.02; direction changes per cell/hr ± SEM, p <

0.0001, n = 5, unpaired t test with Welch’s

correction.

(G) Frequency plot of IN direction changes; dark

gray = Ant1+/+, light gray = Ant1�/�. ****p < 0.0001

and ***p = 0.0006, n = 5, two-way ANOVA with

Sidak’s correction.

In contrast to the clear defects in IN migration, Ant1�/� PN

migration was normal. E14.5 embryos were electroporated

with pCAG-IG and cell positions assayed on E18.5. The loss of

Ant1 did not alter radial migration (Figures S3A and S3B). To

further assay the migration of PNs, we injected 5-ethynyl-2’-de-

oxyuridine (EdU) to pregnant dams on E14.5 and harvested the

embryos on E18.5. Labeling for EdU (E14.5 injections mainly la-

bel outer layer neurons) and Tbr1 (a deeper layer neuronal

marker) showed normal positioning of cortical neurons between

Ant1+/+ and Ant1�/� brains (Figure S4C). Together, these data

indicate that in marked contrast to the non-radial migration of

cortical INs, Ant1 does not appear to affect the radial migration

of PNs.

Loss of Ant1 Alters Centrosome Localization inMigrating InterneuronsTo further examine the migration behaviors disrupting IN migra-

tion in Ant1�/� mutants, we crossed these mice to Dlx5/6CIG

mice to genetically label forebrain GABAergic neurons. Live im-

234 Cell Reports 15, 229–237, April 12, 2016

aging of slices from Ant1�/� and littermate Ant1+/+ controls re-

vealed an �31% decrease in migration rates of GFP+ cells in

the cortex (Figure 4E) and �3.6-fold increases in direction

changes (Figures 4D, 4F, and 4G; see also Movie S7). These

data suggest an impaired ability of Ant1�/� INs to maintain po-

larity. To study this further, we examined centrosome localiza-

tion in MGE explant cultures. In control INs, the centrosome lo-

calizes anterior to the nucleus or in the bleb of the leading

process. Ant1�/� INs displayed markedly aberrant centrosome

positions, posterior to the leading edge of the nucleus, or even

behind the nucleus (Figures 5A and 5B). To confirm this finding,

we also assayed the localization of centrosome after BA.

Similar to the findings in the Ant1�/�, BA-treated migrating

INs also displayed a significant posterior positioning of the

centrosome (Figures 5C and 5D). Taken together with the

increased direction changes seen after either genetic or

biochemical impairment in mitochondrial energetics, these re-

sults suggest that IN polarity is particularly sensitive to mito-

chondrial perturbation.

Page 8: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Figure 5. Loss of Ant1 Causes a Shift in Centrosome Position

(A and C) Sample images of centrosome position in INs show mislocalized

centrosome in Ant�/� INs (A) and BA-treated INs (C) in vitro. Gamma-tubulin,

red; nucleus, blue; cytoplasm, green. Scale bar, 15 mm.

(B and D) Scatterplot of centrosome score of Ant�/� (B) and BA-treated (D) INs

(gray lines, average nuclear length). Ant1+/+: 0.06 ± 0.01; Ant1�/�: 0.02 ± 0.01;

centrosome score ± SEM; p = 0.0005, n = 75 cells each genotype, Mann-

Whitney test, median with interquartile range ± min/max.

DISCUSSION

Our results reveal that migrating INs and PNs display major dif-

ferences in mitochondrial localization. During IN migration, mito-

chondrial localization is highly dynamic, with the highest density

ofmitochondria appearing tomove between the posterior trailing

process, the region anterior to the nucleus, and the cytoplasmic

bleb. In contrast, during PNmigration, mitochondria are primarily

restricted to the region anterior to the nucleus. We also found

that inhibition of OXPHOS drastically decreased the migration

rates of INs, but not PNs. These findings suggest that INs, unlike

radially migrating PNs, are highly dependent on mitochondrial

ATP production. The reduced migratory rates and increased

direction changes by INs also suggest that the maintenance of

polarity is an energetically vulnerable process and is required

for normal IN development. These data link mitochondrial func-

tion to the prenatal development of a critical cerebral cortical

neuronal subpopulation.

Few studies have addressed mitochondrial localization and

trafficking in migrating neurons. Previous work has shown that

Lis1, Tau 1, and DCX, genes that cause defects in radial migra-

tion and IN development, cause mislocalized and altered mito-

chondrial trafficking (Khalaf-Nazzal et al., 2013; Sapir et al.,

2012; Yamada et al., 2009). Although this suggests that defects

in mitochondrial localization may also impact PNs, each of

these genes also regulates microtubule dynamics. Thus, in these

models, it is unclear whether changes in mitochondrial localiza-

tion contribute to the defects in radial migration or whether these

genes have a direct impact on mitochondrial function. Our data

address this issue by investigating mitochondrial localization in

both PN and IN populations and by interfering directly with mito-

chondrial function. Although mitochondrial dysfunction in addi-

tion to other defectsmay contribute to abnormal radial migration,

we provide clear evidence that INs are much more sensitive to

OXPHOS deficits.

Mitochondrial contribution to neuronal metabolism has been

largely studied in the context of the adult nervous system,

focused on how loss of mitochondrial function results in neuro-

degeneration and cell death. Recent data have emphasized

the importance of mitochondrial energetics in basic neurophys-

iology. For example, mitochondrial energetics are essential for

interneuron regulation of gamma oscillations that are themselves

associated with cognitive functions (Kann et al., 2014). However,

few data exist on the earlier developmental requirements for

mitochondrial OXPHOS. Several studies have indicated that

regulation of mitochondrial metabolism impacts neurogenesis

and differentiation (Bertholet et al., 2013; Wang et al., 2014),

but the requirement for OXPHOS during neuronal migration

had not been studied. Surin et al. suggested that glycolysis is

a primary driver of embryonic neuronal metabolism of hippo-

campal cultures (Surin et al., 2012). Since interneurons comprise

only�6% of the neurons in hippocampal cultures (Benson et al.,

1994), it is likely the measurements in this study were primarily

representative of pyramidal neuron metabolism. This lack of

active OXPHOS in embryonic pyramidal cells thus compliments

our findings that PN migration is not impacted by OXPHOS inhi-

bition. Our data clearly show that interneuron migration required

OXPHOS and suggest that distinct neuronal populations have

different metabolic requirements during development.

We found that Ant1mutant INs exhibit changes in centrosome

localization, increased length of the trailing process, and

increased direction changes during IN migration. Mitochondria

have been implicated in centrosome homeostasis in mitotic cells

(Donthamsetty et al., 2014). Additionally, the mislocalization of

centrosomes has also been observed in mice lacking mDia1

and 3, proteins of the formin family that regulate cytoskeletal dy-

namics via Rho-GTPases (Daou et al., 2014). Interestingly, IN

migration is disrupted in these mutants, but radial migration is

not (Shinohara et al., 2012). In this model, focus was on subven-

tricular zonemigration of interneurons to the olfactory bulb; thus,

it is unclear whether there are additional phenotypic similarities

exist between these models. The similarities in our phenotype

and selective effect on INs suggest that the regulation of centro-

somal position and actinomycin contractions within the trailing

process are energetically vulnerable processes and warrant

further investigation.

Patients with ASD, and particularly those with combined ASD,

ID, and epilepsy, commonly have evidence of mitochondrial

dysfunction (Rossignol and Frye, 2012). The manner by which

Cell Reports 15, 229–237, April 12, 2016 235

Page 9: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

mitochondrial dysfunction contributes to these phenotypes is

generally attributed to a deficiency in meeting ongoing neuronal

metabolic demands or increased free radical production result-

ing in cell death. Our data provide clear evidence for a final com-

mon pathway into the pathogenesis of ASD, developmental ep-

ilepsies, and IDs. These clinical phenotypes associated with

mitochondrial disorders may not solely arise from energetic def-

icits or the formation of free radicals during later neuronal func-

tion but may be secondary to abnormal IN development.

EXPERIMENTAL PROCEDURES

Mouse Strains

CD1 or Dlx5/6CIG (Stenman et al., 2003) and Ant1�/� mice on a C57BL6/NJ

(Ronchi et al., 2013) of both sexes were used as indicated. The Institutional

Animal Care and Use Committee at the Children’s Hospital of Philadelphia

(Philadelphia, PA) approved all studies.

Brain Explant and Slice Cultures

Explant and slice cultures were generated from the indicated embryonic day

mouse pups as previously described (Lysko et al., 2011, 2014).

Treatment Protocols

For inhibition of oxidative phosphorylation, explants were cultured for 24 hr in

Dulbecco’smedia (DM) with 35mMglucose. Immediately prior to imaging, me-

dia was exchanged with DM with PBS vehicle, Oligo (Sigma), or BA (Enzo Life

Sciences).We foundastraindifference in response to treatment toBA.CD1cells

were treatedwith 0.5, 2.5, 5, and50mMBA,whileAnt1+/+andAnt1�/� cellswere

treated with 0.5 fM BA. For glucose deprivation and inhibition experiments, ex-

plantswere cultured for 24hr in glucose-freeDMEM (Invitrogen) plusN2supple-

ment (Gibco)with orwithout 10mMsodiumpyruvate (Sigma) or 5mMgalactose

(Sigma), ± 2.5 mMBA. For treatment with 2-DG, explants were cultured for 24 hr

with5mMglucoseDMsupplementedwith 500mM2-DG (Sigma),withorwithout

10 mM sodium pyruvate or 5 mM galactose ± 0.5 mM BA.

Histology and Immunocytochemistry

Brains of E13.5, E16, or E18.5 embryos were processed for histology and

immunohistochemistry as previously described (Lysko et al., 2011). Primary

antibodies used included anti-calbindin D-28k (rabbit; Swant, 1:1,000),

caspase-3 (rabbit; Abcam, 1:500), Ki67 (rabbit; Neomarkers, 1:300), anti-

Tom-20 (rabbit; Santa Cruz Biotechnology, 1:500), and anti-GFP (chicken:

Invitrogen, 1:2,000). Secondary antibodies included goat anti-rabbit-biotin

(Vector Laboratories) followed by Streptavidin/Alexa Fluor 594 (Invitrogen) or

anti-rabbit-Alexa Fluor 594 (Invitrogen) anti-chick-Alexa 488 (Invitrogen), all

at 1:2,000. Nuclei were counterstained with DAPI.

MGE explants were fixed and immunolabeled as previously described (Ly-

sko et al., 2011, 2014) using anti-Tuj1 (rabbit; Neuronal CIII b-tubulin; Covance,

1:1,000), anti gamma-tubulin (mouse; Sigma, 1:200), anti-GFP (chicken; Invi-

trogen, 1:200), or anti-Ant1 (rabbit) as primary antibodies.

Intrauterine Electroporations

Embryos at either E14 or E14.5 were electroporated in utero as previously

described (Nasrallah et al., 2012) with the following constructs: pCAG-IG

(Addgene 11150; 2 mg/ml), pCAG-DsRed (Addgene 11151; 2 mg/ml), pDsRed2-

mito (MitoDsRed;Clontech632421;0.5mg/ml). Formarkingmitochondria in radi-

allymigrating neurons and sliceBA treatments, embryoswere electroporated at

E14 and harvested 48 hr later. For assessing radial migration, embryos were

electroporated at E14.5 and harvested at E18.5.

Live Marking of Mitochondria In Vitro

To image mitochondria in migrating INs, explants from Dlx5/6CIG embryos

were cultured for 24 hr. Prior to imaging, cells were treated with 100 nM Mito-

Tracker Red CMXRos (Invitrogen) for 30 min in Opti-MEM (Invitrogen) with

10 mM glucose. Cells were then rinsed with PBS and supplied with fresh

DM before imaging.

236 Cell Reports 15, 229–237, April 12, 2016

Microscopy

For all experiments, time-lapse images were acquired at indicated intervals for

a minimum of 6 hr with an Olympus Fluoview (FV10i) confocal microscope at

37�C, 5% CO2. Magnifications were as follows: 103 magnification with 23

zoom for treatment protocols at 5-min intervals, and 103 magnification in sli-

ces at 10-min intervals. For higher resolution, 603 magnification was used for

acquiringmitochondrial localization inmigration INs at 10-min intervals. For sli-

ces, z stacks of 10 mm each were taken, capturing the full range of detectable

GFP+ cells or DsRed cells within the slice. Slices were imaged for aminimum of

5 hr. Images of fixed explants cells and slices were taken on an Olympus Fluo-

view (FV10i) confocal microscope at 203 magnification. Mitochondria were

localized within individual cells from 40-mm floating brain slices by collecting

1.5-mm z stacks in using the Olympus Fluoview (FV10i) confocal microscope

at 603 magnification. Mitochondria were labeled with Tom20, and EGFP,

driven by the Dlx5/6 promoter, was detected with the anti-GFP antibody. Im-

ages of calbindin-stained slices and were taken at 53 and 103 magnification

every 5 mm for 15 mm on a Leica CTR600 fluorescent microscope.

Quantification

In all experiments, cells were selected at random using ImageJ’s grid plugin for

all experiments unless indicated otherwise. The color-profiler ImageJ plugin

was used to generate plots of fluorescence units. For fixed cells, mitochondrial

area was calculated by thresholding images using ImageJ’s auto-local thresh-

olding plugin (for invitro culture, Bernsen method, 15 pixels) or by color thresh-

olding images for overlapping green and red pixels (for cells in fixed slices).

Thresholded images were analyzed using the particle analyzer plugin to calcu-

late mitochondrial area in subcellular regions. Distance of migration along the

cortex of the 25 leading cells was measured as a percentage of the distance

between the striatocortical notch and dorsal cortical curve and normalized

to the average distance of wild-type littermates. Relative migration distance

for explants was calculated from the explant edge to the position of the ten

cells that had migrated the furthest. Values were normalized to averages of

Ant1+/+ littermates. Cell migration speed was calculated as previously pub-

lished (Lysko et al., 2011). Leading process orientation was calculated by

designating cells into quadrants based on the orientation of their leading pro-

cesses. Centrosome scores were determined by defining the anterior edge of

the nucleus as zero and centrosomes positioned behind the leading edge of

the nucleus as negative values. Centrosome position was measured from

the posterior of the cell and represented as a percent of total cell length.

Statistics

Prism 6 software was used for all statistical analysis. Data were tested for

normality using either the Kolmogorov-Smirnov test or the D’Agostino and

Pearson omnibus normality test. If the data were not normal, non-parametric

analysis was utilized. p < 0.05 were considered significant. All values are pre-

sented as mean ± SEM unless otherwise indicated.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Supplemental Experimental Procedures,

four figures, and seven movies and can be found with this article online at

http://dx.doi.org/10.1016/j.celrep.2016.03.024.

AUTHOR CONTRIBUTIONS

E.G.L. collected data, processed, and performed analysis. E.G.L., J.A.G., and

S.A.A. contributed to experimental design and wrote the paper. J.A.G. and

S.A.A. contributed equally to the design, execution and interpretation of all

studies. D.C.W. and M.J.N. contributed to pharmacological study design

and experimental interpretation. All authors discussed the results and com-

mented on the manuscript.

ACKNOWLEDGMENTS

This work was supported in part by NIH grants NS21328, NS41850, NS070298

(to D.C.W.), NS46166 (to J.A.G.), Simons Foundation Grant 205844 (to

D.C.W.), and by the RAF Penrose Endowed Chair (to S.A.A.).

Page 10: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Received: January 12, 2015

Revised: February 8, 2016

Accepted: March 4, 2016

Published: March 31, 2016

REFERENCES

Adeva-Andany, M., Lopez-Ojen, M., Funcasta-Calderon, R., Ameneiros-Ro-

drıguez, E., Donapetry-Garcıa, C., Vila-Altesor, M., and Rodrıguez-Seijas, J.

(2014). Comprehensive review on lactate metabolism in human health. Mito-

chondrion 17, 76–100.

Bellion, A., Baudoin, J.P., Alvarez, C., Bornens, M., and Metin, C. (2005). Nu-

cleokinesis in tangentially migrating neurons comprises two alternating

phases: forward migration of the Golgi/centrosome associated with centro-

some splitting and myosin contraction at the rear. J. Neurosci. 25, 5691–5699.

Benson, D.L., Watkins, F.H., Steward, O., and Banker, G. (1994). Characteriza-

tion of GABAergic neurons in hippocampal cell cultures. J. Neurocytol. 23,

279–295.

Bertholet, A.M., Millet, A.M., Guillermin, O., Daloyau, M., Davezac, N., Miquel,

M.C., and Belenguer, P. (2013). OPA1 loss of function affects in vitro neuronal

maturation. Brain 136, 1518–1533.

Cheng, A., Wan, R., Yang, J.L., Kamimura, N., Son, T.G., Ouyang, X., Luo, Y.,

Okun, E., and Mattson, M.P. (2012). Involvement of PGC-1a in the formation

and maintenance of neuronal dendritic spines. Nat. Commun. 3, 1250.

Chevallier, J.A., Von Allmen, G.K., and Koenig, M.K. (2014). Seizure semiology

and EEG findings in mitochondrial diseases. Epilepsia 55, 707–712.

Daou, P., Hasan, S., Breitsprecher, D., Baudelet, E., Camoin, L., Audebert, S.,

Goode, B.L., and Badache, A. (2014). Essential and nonredundant roles for

Diaphanous formins in cortical microtubule capture and directed cell migra-

tion. Mol. Biol. Cell 25, 658–668.

Donthamsetty, S., Brahmbhatt, M., Pannu, V., Rida, P.C., Ramarathinam, S.,

Ogden, A., Cheng, A., Singh, K.K., and Aneja, R. (2014). Mitochondrial genome

regulates mitotic fidelity by maintaining centrosomal homeostasis. Cell Cycle

13, 2056–2063.

Golden, J.A., Zitz, J.C., McFadden, K., and Cepko, C.L. (1997). Cell migration

in the developing chick diencephalon. Development 124, 3525–3533.

Graham, B.H.,Waymire, K.G., Cottrell, B., Trounce, I.A., MacGregor, G.R., and

Wallace, D.C. (1997). A mouse model for mitochondrial myopathy and cardio-

myopathy resulting from a deficiency in the heart/muscle isoform of the

adenine nucleotide translocator. Nat. Genet. 16, 226–234.

Henderson, P.J., and Lardy, H.A. (1970). Bongkrekic acid. An inhibitor of the

adeninenucleotide translocaseofmitochondria. J. Biol. Chem.245, 1319–1326.

Kann, O., Papageorgiou, I.E., and Draguhn, A. (2014). Highly energized inhib-

itory interneurons are a central element for information processing in cortical

networks. J. Cereb. Blood Flow Metab. 34, 1270–1282.

Khalaf-Nazzal, R., Bruel-Jungerman, E., Rio, J.P., Bureau, J., Irinopoulou, T.,

Sumia, I., Roumegous, A., Martin, E., Olaso, R., Parras, C., et al. (2013). Organ-

elle and cellular abnormalities associated with hippocampal heterotopia in

neonatal doublecortin knockout mice. PLoS ONE 8, e72622.

Kimura, T., and Murakami, F. (2014). Evidence that dendritic mitochondria

negatively regulate dendritic branching in pyramidal neurons in the neocortex.

J. Neurosci. 34, 6938–6951.

Kulka, R.G., and Cooper, C. (1962). The action of oligomycin on the inorganic

orthophosphate-adenosine triphosphate and adenosine diphosphate-adeno-

sine triphosphate exchange reactions of digitonin particles. J. Biol. Chem. 237,

936–939.

Lee, J., Schriner, S.E., andWallace, D.C. (2009). Adenine nucleotide transloca-

tor 1 deficiency increases resistance of mouse brain and neurons to excito-

toxic insults. Biochim. Biophys. Acta 1787, 364–370.

Levy, S.E., Chen, Y.S., Graham, B.H., and Wallace, D.C. (2000). Expression

and sequence analysis of the mouse adenine nucleotide translocase 1 and 2

genes. Gene 254, 57–66.

Lysko, D.E., Putt, M., and Golden, J.A. (2011). SDF1 regulates leading process

branching and speed of migrating interneurons. J. Neurosci. 31, 1739–1745.

Lysko, D.E., Putt, M., and Golden, J.A. (2014). SDF1 reduces interneuron lead-

ing process branching through dual regulation of actin and microtubules.

J. Neurosci. 34, 4941–4962.

Marın, O. (2012). Interneuron dysfunction in psychiatric disorders. Nat. Rev.

Neurosci. 13, 107–120.

Marın, O., Valdeolmillos, M., and Moya, F. (2006). Neurons in motion: same

principles for different shapes? Trends Neurosci. 29, 655–661.

Marroquin, L.D., Hynes, J., Dykens, J.A., Jamieson, J.D., and Will, Y. (2007).

Circumventing the Crabtree effect: replacing media glucose with galactose in-

creases susceptibility of HepG2 cells to mitochondrial toxicants. Toxicol. Sci.

97, 539–547.

Nasrallah, M.P., Cho, G., Simonet, J.C., Putt, M.E., Kitamura, K., and Golden,

J.A. (2012). Differential effects of a polyalanine tract expansion in Arx on neural

development and gene expression. Hum. Mol. Genet. 21, 1090–1098.

Noctor, S.C., Martınez-Cerdeno, V., Ivic, L., and Kriegstein, A.R. (2004).

Cortical neurons arise in symmetric and asymmetric division zones and

migrate through specific phases. Nat. Neurosci. 7, 136–144.

Polleux, F., Whitford, K.L., Dijkhuizen, P.A., Vitalis, T., and Ghosh, A. (2002).

Control of cortical interneuron migration by neurotrophins and PI3-kinase

signaling. Development 129, 3147–3160.

Ronchi, J.A., Figueira, T.R., Ravagnani, F.G., Oliveira, H.C., Vercesi, A.E., and

Castilho, R.F. (2013). A spontaneous mutation in the nicotinamide nucleotide

transhydrogenase gene of C57BL/6J mice results in mitochondrial redox ab-

normalities. Free Radic. Biol. Med. 63, 446–456.

Rossignol, D.A., and Frye, R.E. (2012). Mitochondrial dysfunction in autism

spectrum disorders: a systematic review and meta-analysis. Mol. Psychiatry

17, 290–314.

Sapir, T., Frotscher, M., Levy, T., Mandelkow, E.M., and Reiner, O. (2012).

Tau’s role in the developing brain: implications for intellectual disability.

Hum. Mol. Genet. 21, 1681–1692.

Schaefer, A.M., Taylor, R.W., Turnbull, D.M., and Chinnery, P.F. (2004). The

epidemiology of mitochondrial disorders–past, present and future. Biochim.

Biophys. Acta 1659, 115–120.

Shinohara, R., Thumkeo, D., Kamijo, H., Kaneko, N., Sawamoto, K., Wata-

nabe, K., Takebayashi, H., Kiyonari, H., Ishizaki, T., Furuyashiki, T., and Naru-

miya, S. (2012). A role for mDia, a Rho-regulated actin nucleator, in tangential

migration of interneuron precursors. Nat. Neurosci. 15, 373–380, S371–S372.

Stenman, J., Toresson, H., and Campbell, K. (2003). Identification of two

distinct progenitor populations in the lateral ganglionic eminence: implications

for striatal and olfactory bulb neurogenesis. J. Neurosci. 23, 167–174.

Surin, A.M., Khiroug, S., Gorbacheva, L.R., Khodorov, B.I., Pinelis, V.G., and

Khiroug, L. (2012). Comparative analysis of cytosolic and mitochondrial ATP

synthesis in embryonic and postnatal hippocampal neuronal cultures. Front.

Mol. Neurosci. 5, 102.

Valenti, D., de Bari, L., De Filippis, B., Henrion-Caude, A., and Vacca, R.A.

(2014). Mitochondrial dysfunction as a central actor in intellectual disability-

related diseases: an overview of Down syndrome, autism, Fragile X and Rett

syndrome. Neurosci. Biobehav. Rev. 46, 202–217.

Wang, L., Ye, X., Zhao, Q., Zhou, Z., Dan, J., Zhu, Y., Chen, Q., and Liu, L.

(2014). Drp1 is dispensable for mitochondria biogenesis in induction to plurip-

otency but required for differentiation of embryonic stem cells. StemCells Dev.

23, 2422–2434.

Yamada, M., Yoshida, Y., Mori, D., Takitoh, T., Kengaku, M., Umeshima, H.,

Takao, K., Miyakawa, T., Sato, M., Sorimachi, H., et al. (2009). Inhibition of cal-

pain increases LIS1 expression and partially rescues in vivo phenotypes in a

mouse model of lissencephaly. Nat. Med. 15, 1202–1207.

Cell Reports 15, 229–237, April 12, 2016 237

Page 11: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Cell Reports, Volume 15

Supplemental Information

Differential Mitochondrial Requirements

for Radially and Non-radially Migrating Cortical

Neurons: Implications for Mitochondrial Disorders

Erika G. Lin-Hendel, Meagan J. McManus, Douglas C. Wallace, Stewart A.Anderson, and Jeffrey A. Golden

Page 12: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Detailed methods:

Mouse Strains CD1, or Dlx5/6-CIG mice (Stenman et al., 2003), were used for mitochondrial localization and pharmacological experiments. Ant1+/+, Ant1-/- mice on a C57BL6/NJ (Ronchi et al., 2013) strain were used as indicated. Both sexes were used for experiments. The Institutional Animal Care and Use Committee at the Children’s Hospital of Philadelphia, Philadelphia, PA, approved all studies. Brain explant culture For all in vitro experiments, explants were generated from at embryonic day 14.5. Embryonic brains were dissected on ice cold HBSS (Sigma), embedded in 4% low-melt agarose (Lonza) and sliced into 250 µm thick sections. The MGE was microdissected in DFS (DMEM:F12; Invitrogen, with 10% fetal bovine serum, 40 mM Glucose, 40µM L-Glutamine) and cut into ~200 µm cubes, and were placed in a gel of 35% acidic collagen (0.0035 N acetic acid in rat tail collagen; BD Pharmogen), 50% matrigel (BD Pharmogen), 15% DM (DM; glucose free DMEM (Invitrogen), N2 supplement (Gibco), +/- Glucose as indicated) and 5 mM HEPES. Explants and gel incubated at for 30 minutes at 37 °C 5% CO2 to set, and DM medium added. Explants were continually cultured in DM with glucose as indicated. Brain slice culture Brains were dissected on ice cold HBSS (Sigma), embedded in 4% low-melt agarose (Lonza) and sliced into 250 µm thick sections on a Lieca VT1200S vibratome, and placed on filter inserts (Millipore). Slices were covered with a stiff gel of rat tail collagen, 3.75% Sodium Bicarbonate, and 25% DM and incubated at 37 °C 5% CO2 in DFS for 20 minutes to recover. Slices were then placed in 5mM glucose DM +/- BA as indicated and imaged as described below. Treatment protocols For inhibition of oxidative phosphorylation, explants were cultured for 24 hours in DM with 35 mM glucose. Immediately prior to imaging, media was exchanged with DM with PBS vehicle, Oligomycin (Sigma), or Bongkrekic Acid (Enzo Life Sciences). We found a strain difference in response to treatment to BA. CD1 cells were treated with 0.5, 2.5, 5, and 50 µM BA, while Ant1+/+ and Ant1-/- were treated with 0.5 fM BA. For glucose deprivation and inhibition experiments, explants were cultured for 24 hours in glucose free DMEM (Invitrogen) plus N2 supplement (Gibco) with or without 10 mM sodium pyruvate (Sigma) or 5 mM Galactose (Sigma), +/- 2.5 µM BA. For treatment with 2-DG, explants were cultured for 24 hours with 5 mM glucose DM supplemented with 500 µM 2-DG (Sigma), with or without 10 mM sodium pyruvate or 5 mM galactose +/- 0.5 µM BA. Histology and immunocytochemistry Brains of E13.5, E16, or E18.5 embryos were dissected on ice cold Hanks and drop fixed into 4% PFA overnight. A Leica VT1200S vibratome was used to cut floating sections of E13.5 (40 µm), E16.5, or E18.5 brains (50 µm thick). Sections were blocked in 5% NGS, PBS-T and stained with anti-Calbindin D-28k (rabbit; Swant, 1:1000), Caspase-3 (rabbit; Abcam, 1:500), Ki67 (rabbit; Neomarkers, 1:300, 5 min antigen retrieval with 1mM EDTA at 42oC), anti-Tom-20 (rabbit; Santa Cruz, 1:500), anti-GFP (chicken: Invitrogen, 1:2000). Slices were incubated in primary antibody for 24-48 hours, washed and stained with secondary antibody for 1 hour at room temperature (goat anti-rabbit-biotin; Vector Laboratories followed by Streptavidin-Alexa 594; Invitrogen, or anti-rabbit-Alexa 594; Invitrogen anti-chick-Alexa 488; Invitrogen all at 1:2000). Nuclei were counterstained with DAPI. MGE explants were fixed in methanol for 20 minutes at -20 °C for gamma tubulin staining, or for 12 minutes in 4% paraformaldehyde and PHEM buffer at 37 °C. After washing in 1x PBS, explants were stored in 1x PBS at 4 °C until staining. For staining, explants were treated with 1:50 collagenase (Roche) for 15 minutes, permeabilized for 15 minutes with 0.04% triton. Explants were blocked with 10% NGS for 1 hour at RT. Explants were incubated overnight at 4ºC with primary antibodies anti-Tuj1 (rabbit; Neuronal CIII b-tubulin; Covance, 1:1000), anti gamma-tubulin (mouse; 1:200 Sigma), anti-GFP (chicken Invitrogen 1:200), or anti-Ant1 (rabbit). The Ant1 antibody was generated by Covance® using the oligopeptide derived from the N-terminal sequences of Ant1 NH2-MGDQALSFLKDFLAG injected into rabbits (Graham et al., 1997). The antibody specificity was confirmed by western blot (Figure S3E). Intrauterine electroporations Embryos at either E14, or E14.5 were electroporated in utero as previously published (Nasrallah et al., 2012) with the following constructs: pCAG-IG (Addgene 11150; 2 µg/uL), pCAG-DsRed (Addgene 11151; 2 ug/uL), pDsRed2-mito (MitoDsRed; Clontech 632421; 0.5 µg/uL). For marking mitochondria in radially migrating neurons, and slice BA treatments, embryos were electroporated at E14 and harvested 48 hrs later. For assessing radial migration, embryos were electroporated at E14.5 and harvested at E18.5. Live marking of mitochondria in vitro

Page 13: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

To image mitochondria in migrating INs, explants from Dlx5/6CIG embryos were cultured for 24 hrs. Prior to imaging, cells were treated with 100 nM MitoTracker® Red CMXRos (Invitrogen) for 30 minutes in Opti-MEM (Invitrogen) with 10 mM glucose. Cells were then rinsed with PBS and supplied with fresh DM before imaging. Microscopy For all experiments, time lapse images were acquired at indicated intervals for a minimum of 6 hours with an Olympus Fluoview (FV10i) confocal microscope at 37°C, 5% CO2. Magnifications were as follows; 10X magnification with 2X zoom for treatment protocols in 5-minute intervals, and 10X magnification in slices in 10-minute intervals. For higher resolution, 60X magnification was used for acquiring mitochondrial localization in migration INs at 10-minute intervals. For slices, Z-stacks of 10 µm each were taken, capturing the full range of detectable GFP+ cells or DsRed cells within the slice. Slices were imaged for a minimum of 5 hrs. Images of fixed explants cells and slices were taken on an Olympus Fluoview (FV10i) confocal microscope at 20x magnification. Mitochondria were localized within individual cells from 40µm floating brain slices by collecting 1.5µm z-stacks in using the Olympus Fluoview (FV10i) confocal microscope at 60x magnification. Mitochondria were labeled with Tom20 and eGFP, driven by the Dlx5/6 promoter, was detected with the anti-GFP antibody. Images of calbindin stained slices and were taken at 5X and 10X magnification every 5 µm for 15 µm on a Leica CTR600 fluorescent microscope. Quantification In all experiments, cells were selected at random using ImageJ’s grid plugin for all experiments unless indicated otherwise. Mitochondrial localization For mitochondrial localization live imaging experiments, all INs that were in focus for a cell migration cycle, had Mitotracker® uptake, and were healthy were counted. Unhealthy cells were considered those that had halted migration and displayed leaky Mitotracker® signal, and those that died over the course of imaging. For radially migrating cells, all cells in the cortical plate that were in focus and expressing MitoDsRed were counted. The color-profiler ImageJ plugin was used to generate plots of fluorescence units. For fixed cells, mitochondrial area was calculated by thresholding images using ImageJ’s auto-local thresholding plugin (for invitro culture, Bernsen method, 15 pixels), or by color thresholding images for overlapping green and red pixels (for cells in fixed slices). Thresholded images were analyzed using the particle analyzer plugin to calculate mitochondrial area in subcellular regions. Distance of migration of calbindin positive cells in cortex. The distance along the cortex of the 25 leading cells was measured as a percentage of the distance between the striato-cortical notch and dorsal cortical curve, and normalized to the average distance of wild type littermates. Relative Migration Distance. Explants were cultured for 16 hours from Ant1+/+ and Ant1-/- individuals from three separate litters. The distance from the explant edge to the position of the 10 cells that had migrated the furthest was measured. Values were normalized to averages of Ant1+/+ littermates. A minimum of 5 separate explants were analyzed for each experiment, and a minimum of 50 cells were measured from each individual explant. Cell migration characteristics. Cell migration speed was calculated as previously published (Lysko et al., 2011). For both explant and slice imaging, cells were selected at random and followed during the live imaging process using ImageJ Manual Tracking plugin. For in vitro experiments, a minimum of 25 cells per n were followed. For slice experiments, a minimum of 15 PNs and 20 INs were tracked per individual. For radially migrating cells, cells that were not in the cortical plate were excluded. For assessing tangential migration in slice culture for BA treatment, GFP+ cells that were purely in the radial phase of their migration were excluded. Direction changes were defined as any change in direction greater than 90 degrees. Both direction changes and trailing process length were manually measured. For Ant1 Dlx-GFPCIG experiments, a minimum of 20 cells per individual were selected, and migration rates were normalized to the average rate of littermate controls. Leading process orientation. 25 cells per individual were selected at random as previously described. Cells were designated into quadrants for the orientation of their leading processes (Figure 3H, insert) Q1, dorsal, Q2, pial; Q3, ventral; Q4, ventricular orientation. Centrosome score. The anterior edge of the nucleus was defined as zero, with centrosomes positioned behind the leading edge of the nucleus as negative values. Centrosome position was measured from the posterior of the cell and represented as a percent of total cell length. Statistics. Prism 6 software was used for all statistical analysis. Data were tested for normality using either the K-S test or the D’Agostino & Pearson omnibus normality test. If the data were not normal, non-parametric analysis was utilized. p < 0.05 were considered significant. All values are represented as mean ± SEM unless otherwise indicated.

Page 14: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Figure S1, relates to Figure 1 and Movie S1

(A) Neurons were visually scored as matching or non-matching to morphology patterns seen in vitro with mitochondrial clustering in front of the nucleus, within the bleb, or the trailing process. (B-D) Quantification of clustering of mitochondria in subcellular locations. Region I: trailing process (TP); II: overlapping nucleus (Nuc); III: 5 µm anterior to nucleus (5 µm AN); IV: cytoplasmic bleb (bleb); V: leading process (LP). Similar to in vitro data, clustering varied between IN morphologies. IN morphology 1 clustered in III, *p ≤ 0.029. IN morphology 2, clustered in IV, *p ≤ 0.022 The clustering of mitochondria within the trailing process of IN morphology 3 was not as distinct compared to in vitro data. Clustering was higher in region I compared to region II (p < 0.0001), most mitochondria were clustered in region III (p = 0.04). Bars represent median with Tukey distribution, dots represent calculations beyond 1.5 times interquartile range of the percent total mitochondrial area (%TMA) normalized to region’s percent of total cell area (%TCA). n ≥ 50 cells each morphology from 5 different individuals. Freidman’s test with Dunn’s correction.

Page 15: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Figure S2, relates to Figure 2

(A) Oligomycin treatment reduced IN migration rates. (Oligo: p <0.0001, N = 3 independent cultures, 150 cells each, Mann-Whitney). Mean distance for Oligomycin concentrations of 0 µM: 0.31 ± 0.01 and 0.02 µM: 0.068 ± 0.005, µm/min ± SEM. (B) Representative phase image of IN cells treated with Oligomycin. White arrows = extended trailing processes. (C) IN migration is not dependent on glycolysis, as OXPHOS substrate pyruvate (Pyr) is sufficient to rescue decreased migration from glucose deprivation, and inhibition with 2-Deoxyglucose (2-DG). Pyr = pyruvate, Gluc = glucose, 2-DG = 2-deoxyglucose, BA = bongkrekic acid. p ≤ 0.001 as indicated, n = 5 independent experiments, >75 cells each, Kruskall-Wallis with Dunn’s correction. Data not shown for Galactose.

Page 16: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Figure S3, relates to Figure 3 Ant1 is expressed in MGE derived INs.

(A) Ant1 expression is detectable in migrating Dlx5/6CIG INs in explant cultures. Scale bar = 20 µm. (B) Western blot of whole brain lysate showing specificity of Ant1 antibody.

Page 17: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Figure S4, Relates to Figure 4 Ant1 -/- cortices display normal radial migration

(A) Representative max projection of confocal images of E14.5 cortices electroporated in utero with pCAG-IRES-GFP, marking radially migrating PNs. After 4 days, radial migration was assessed, and was normal in the cortices of Ant1-/- embryos. (B) Quantification of percent total GFP+ cells in defined bins dividing cortex: ventricular and intermediate zones, and 8 equally sized divisions of the cortical plate (VZ: IZ; 1-8). p > 0.5, n = 5 individuals for both genotypes, two-way ANOVA. Scale bar = 250 µm. (C) Representative coronal sections from E18.5 Ant1+/+ and Ant1-/- brains at similar anterior posterior levels showing Tbr1 (red) labeled cells normally positioned in the deeper cortical layers and neurons born on E14.5 (injection time for EdU, labeled in green) normally positioned in the outer cortical layers and above the Tbr1 labeled cells. All nuclei stained with DAPI (blue). No difference was observed (n=3, scale bar=500 µm)

Page 18: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Supplementary Movie 1 Relates to Figure 1

Dynamic mitochondrial localization in migrating INs within whole brain slices. Confocal immunofluorescence images of mitochondria in migrating INs in situ displaying varying localization patterns according to morphology. (IN; 1-3) morphologies displayed during migration. Outline of cell (yellow dotted line), Cytosol (GFP), mitochondria within GFP cells (white), mitochondria (Tom20), nuclei (DAPI). Scale bar = 10 µm.

Page 19: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Supplementary Movie 2 Relates to Figure 1

Dynamic mitochondrial localization in dissociated migrating INs. Time-lapse imaging of a migrating interneurons treated with Mitotracker® Red CMXRos. 1 frame is 10 minutes, 3 frames per second (fps); scale bar, 10 µm. Relative fluorescence of mitochondrial signal and cytosolic GFP represented in ImageJ color plot profile.

Page 20: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Supplementary Movie 3 Relates to Figure 1

Restricted mitochondrial localization in migrating PNs. Time-lapse imaging of a radially migrating pyramidal neuron expressing pCAG-IG and MitoDsRed in slice culture. 1 frame is 10 minutes, 3 fps; scale bar, 10 µm. Relative fluorescence of mitochondrial signal and cytosolic GFP represented in ImageJ color plot profile.

Page 21: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Supplementary Movie 4 Relates to Figure 2

Oligomycin reduces IN migration in vitro. Time-lapse imaging of migrating interneuron in vitro treated with vehicle (PBS), or Oligomycin (0.02 µM). 1 frame is 5 minutes, 3 fps; scale bar, 40 µm.

Page 22: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Supplementary Movie 5 Relates to Figure 2

Bongkrekic Acid reduces IN migration in vitro. Time-lapse imaging of migrating interneuron in vitro treated with vehicle (PBS), or BA (0 µM, 2.5 µM, 5 µM). 1 frame is 5 minutes, 3 fps; scale bar, 40 µm.

Page 23: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Supplementary Movie 6 Relates to Figure 2

Abnormal tangential but normal radial migration in slice cultures treated with Bongkrekic Acid. Slice cultures treated with BA (20 µM right) versus vehicle alone (left) display abnormal interneuron migration, and normal radial migration in situ. 1 frame is 10 minutes, 3 fps; scale bar, 250 µm.

Page 24: Differential Mitochondrial Requirements for Radially and ... · these two neuronal cell populations during development. Oxidative Phosphorylation Is Necessary for Normal IN Migration,

Supplementary Movie 7 Relates to Figure 4

Ant1-/- INs display abnormal migration in situ Time-lapse imaging of Ant1 -/- and Ant1+/+ GFP+ migrating interneurons in slice culture. 1 frame is 10 minutes, 3 fps; scale bar, 250 µm.