genetic recovery of erbb4 in adulthood partially restores ... · genetic recovery of erbb4 in...

6
Genetic recovery of ErbB4 in adulthood partially restores brain functions in null mice Hongsheng Wang a,1 , Fang Liu b,1 , Wenbing Chen a,c,1 , Xiangdong Sun d , Wanpeng Cui a , Zhaoqi Dong a , Kai Zhao b , Hongsheng Zhang a , Haiwen Li a , Guanglin Xing a , Erkang Fei c , Bing-Xing Pan c , Bao-Ming Li c , Wen-Cheng Xiong a,e , and Lin Mei a,e,2 a Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106; b Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912; c Institute of Life Science, Nanchang University, 330031 Nanchang, China; d School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, 510260 Guangzhou, Guangdong, China; and e Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106 Edited by Richard L. Huganir, The Johns Hopkins University School of Medicine, Baltimore, MD, and approved November 1, 2018 (received for review July 1, 2018) Neurotrophic factor NRG1 and its receptor ErbB4 play a role in GABAergic circuit assembly during development. ErbB4 null mice possess fewer interneurons, have decreased GABA release, and show impaired behavior in various paradigms. In addition, NRG1 and ErbB4 have also been implicated in regulating GABAergic transmission and plasticity in matured brains. However, current ErbB4 mutant strains are unable to determine whether phenotypes in adult mutant mice result from abnormal neural development. This important question, a glaring gap in understanding NRG1ErbB4 function, was addressed by using two strains of mice with temporal control of ErbB4 deletion and expression, respectively. We found that ErbB4 deletion in adult mice impaired behavior and GABA release but had no effect on neu- ron numbers and morphology. On the other hand, some deficits due to the ErbB4 null mutation during development were alleviated by restoring ErbB4 expression at the adult stage. Together, our results indicate a critical role of NRG1ErbB4 signaling in GABAergic trans- mission and behavior in adulthood and suggest that restoring NRG1ErbB4 signaling at the postdevelopmental stage might benefit relevant brain disorders. ErbB4 | adulthood | GABA | treatment | schizophrenia S chizophrenia (SZ) is a disabling mental disorder that affects 1% of the population worldwide (1). It alters basic brain processes of perception, emotion, and judgment to cause hallu- cinations, delusions, thought disorder, anhedonia, and cognitive deficits. Despite extensive efforts to study its pathophysiological mechanisms, SZ remains one of the least understood brain dis- orders. Recent identification of SZ susceptibility genes and studies of their functions have begun to shed light on its pathophysiology. Neuregulin 1 (NRG1) is an EGF-domaincontaining trophic fac- tor that acts by activating ErbB tyrosine kinases including ErbB4. NRG1ErbB4 signaling is critical for assembling GABAergic cir- cuits. Mutant mice lacking NRG1 or ErbB4 display deficits in inter- neuron migration, axon and dendrite development of interneurons, and synaptogenesis onto and by interneurons (210). Both NRG1 and ErbB4 are expressed in the adult brain. NRG1 is produced mainly in neurons in an activity-dependent manner (11, 12). On the other hand, 99% of ErbB4-positive neurons in the adult cortex and hippocampus are GABAergic (4, 1317), the majority of which express parvalbumin (PV) (4, 13, 15, 16). In adult animals, NRG1ErbB4 signaling could promote GABAergic transmission and thus control pyramidal neuron activity (1724). It was proposed that NRG1ErbB4 signaling serves as a homeostatic mechanism to control the excitationinhibition (E-I) balance (25, 26). Interestingly, NRG1 and ErbB4 are SZ risk genes in diverse populations based on family trio studies, case-controlled associ- ation studies, and meta-analysis studies [see review by Mei and Nave (25)]. However, their polymorphism did not reach genome- wide significance in a recent genome-wide association study of SZ (27), perhaps as a result of allelic heterogeneity at NRG1 and ErbB4 loci, the existence of haplotypes (2832), and/or pop- ulation stratification (3336). The following evidence supports the notion that NRG1 and ErbB4 alteration contributes to pathophysiological mechanisms of SZ. First, NRG1 and ErbB4 variants have been shown to be associated with reduced volume and decreased activation of brain regions and cognitive pheno- types (3739) and with responses to antipsychotics treatment (4042). Second, altered NRG1 or ErbB4 levels were detected in postmortem brain samples and peripheral blood of SZ patients (4345). Third, mutating NRG1 and ErbB4 or altering their levels in mice could recapitulate SZ-related endophenotypes (3, 18, 24, 4648). Finally, recent meta-analyses (4951), including one from 2017 on >16,000 schizophrenic patients and >20,000 controls (49), identify NRG1 and ErbB4 as risk genes for SZ. In- terestingly, SNP rs7598440 of ErbB4 could predict cortical or ce- rebrospinal fluid GABA concentration in healthy human subjects (52, 53), in agreement with critical roles of ErbB4 in the develop- ment and function of the GABA circuitry from mouse studies. A critical question in understanding how abnormal NRG1ErbB4 signaling alters brain functions is whether it also involves the E-I imbalance in adulthood. A related question is whether SZ-associated endophenotypes caused by ErbB4 deficiency in early development could be diminished by restoring ErbB4 ex- pression in adult animals. These questions require temporal control of ErbB4 mutation and expression. In this paper, we developed genetic approaches; in inducible knockout (iKO) Significance NRG1ErbB4 signaling is implicated in GABAergic circuit as- sembly during development and GABAergic transmission at adulthood. However, it is unclear whether phenotypes in the adult stage in ErbB4 mutant mice result from abnormal neural development. By using two strains of mice with temporal control of ErbB4 deletion and expression, we demonstrate that ErbB4 deletion in adult mice impaired behavior and GABA re- lease, whereas deficits due to ErbB4 null mutation during de- velopment were alleviated by restoring ErbB4 expression at the adult stage. Together, our results indicate that NRG1ErbB4 signaling at adulthood is critical to GABAergic transmission and behavior and suggest that restoring NRG1ErbB4 signaling at the postdevelopmental stage might benefit relevant brain disorders. Author contributions: H.W., F.L., W. Chen, B.-M.L., W.-C.X., and L.M. designed research; H.W., F.L., W. Chen, X.S., W. Cui, Z.D., K.Z., H.Z., H.L., G.X., E.F., and B.-X.P. performed research; H.W., F.L., and W. Chen analyzed data; and H.W., F.L., W. Chen, and L.M. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. Published under the PNAS license. 1 H.W., F.L., and W. Chen contributed equally to this work. 2 To whom correspondence should be addressed. Email: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1811287115/-/DCSupplemental. Published online November 29, 2018. www.pnas.org/cgi/doi/10.1073/pnas.1811287115 PNAS | December 18, 2018 | vol. 115 | no. 51 | 1310513110 NEUROSCIENCE Downloaded by guest on December 6, 2020

Upload: others

Post on 22-Aug-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Genetic recovery of ErbB4 in adulthood partially restores ... · Genetic recovery of ErbB4 in adulthood partially restores brain functions in null mice Hongsheng Wanga,1, Fang Liub,1,

Genetic recovery of ErbB4 in adulthood partiallyrestores brain functions in null miceHongsheng Wanga,1, Fang Liub,1, Wenbing Chena,c,1, Xiangdong Sund, Wanpeng Cuia, Zhaoqi Donga, Kai Zhaob,Hongsheng Zhanga, Haiwen Lia, Guanglin Xinga, Erkang Feic, Bing-Xing Panc, Bao-Ming Lic, Wen-Cheng Xionga,e,and Lin Meia,e,2

aDepartment of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106; bDepartment of Neuroscience and RegenerativeMedicine, Medical College of Georgia, Augusta University, Augusta, GA 30912; cInstitute of Life Science, Nanchang University, 330031 Nanchang, China;dSchool of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, 510260 Guangzhou, Guangdong, China; and eLouisStokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106

Edited by Richard L. Huganir, The Johns Hopkins University School of Medicine, Baltimore, MD, and approved November 1, 2018 (received for review July1, 2018)

Neurotrophic factor NRG1 and its receptor ErbB4 play a role inGABAergic circuit assembly during development. ErbB4 null micepossess fewer interneurons, have decreased GABA release, and showimpaired behavior in various paradigms. In addition, NRG1 and ErbB4have also been implicated in regulating GABAergic transmission andplasticity in matured brains. However, current ErbB4 mutant strainsare unable to determine whether phenotypes in adult mutant miceresult from abnormal neural development. This important question, aglaring gap in understanding NRG1–ErbB4 function, was addressedby using two strains of mice with temporal control of ErbB4 deletionand expression, respectively. We found that ErbB4 deletion in adultmice impaired behavior and GABA release but had no effect on neu-ron numbers and morphology. On the other hand, some deficits dueto the ErbB4 null mutation during development were alleviated byrestoring ErbB4 expression at the adult stage. Together, our resultsindicate a critical role of NRG1–ErbB4 signaling in GABAergic trans-mission and behavior in adulthood and suggest that restoring NRG1–ErbB4 signaling at the postdevelopmental stage might benefitrelevant brain disorders.

ErbB4 | adulthood | GABA | treatment | schizophrenia

Schizophrenia (SZ) is a disabling mental disorder that affects∼1% of the population worldwide (1). It alters basic brain

processes of perception, emotion, and judgment to cause hallu-cinations, delusions, thought disorder, anhedonia, and cognitivedeficits. Despite extensive efforts to study its pathophysiologicalmechanisms, SZ remains one of the least understood brain dis-orders. Recent identification of SZ susceptibility genes and studiesof their functions have begun to shed light on its pathophysiology.Neuregulin 1 (NRG1) is an EGF-domain–containing trophic fac-

tor that acts by activating ErbB tyrosine kinases including ErbB4.NRG1–ErbB4 signaling is critical for assembling GABAergic cir-cuits. Mutant mice lacking NRG1 or ErbB4 display deficits in inter-neuron migration, axon and dendrite development of interneurons,and synaptogenesis onto and by interneurons (2–10). Both NRG1and ErbB4 are expressed in the adult brain. NRG1 is producedmainly in neurons in an activity-dependent manner (11, 12). On theother hand, 99% of ErbB4-positive neurons in the adult cortex andhippocampus are GABAergic (4, 13–17), the majority of whichexpress parvalbumin (PV) (4, 13, 15, 16). In adult animals, NRG1–ErbB4 signaling could promote GABAergic transmission and thuscontrol pyramidal neuron activity (17–24). It was proposed thatNRG1–ErbB4 signaling serves as a homeostatic mechanism tocontrol the excitation–inhibition (E-I) balance (25, 26).Interestingly, NRG1 and ErbB4 are SZ risk genes in diverse

populations based on family trio studies, case-controlled associ-ation studies, and meta-analysis studies [see review by Mei andNave (25)]. However, their polymorphism did not reach genome-wide significance in a recent genome-wide association study ofSZ (27), perhaps as a result of allelic heterogeneity at NRG1 andErbB4 loci, the existence of haplotypes (28–32), and/or pop-ulation stratification (33–36). The following evidence supports

the notion that NRG1 and ErbB4 alteration contributes topathophysiological mechanisms of SZ. First, NRG1 and ErbB4variants have been shown to be associated with reduced volumeand decreased activation of brain regions and cognitive pheno-types (37–39) and with responses to antipsychotics treatment(40–42). Second, altered NRG1 or ErbB4 levels were detected inpostmortem brain samples and peripheral blood of SZ patients(43–45). Third, mutating NRG1 and ErbB4 or altering theirlevels in mice could recapitulate SZ-related endophenotypes (3,18, 24, 46–48). Finally, recent meta-analyses (49–51), includingone from 2017 on >16,000 schizophrenic patients and >20,000controls (49), identify NRG1 and ErbB4 as risk genes for SZ. In-terestingly, SNP rs7598440 of ErbB4 could predict cortical or ce-rebrospinal fluid GABA concentration in healthy human subjects(52, 53), in agreement with critical roles of ErbB4 in the develop-ment and function of the GABA circuitry from mouse studies.A critical question in understanding how abnormal NRG1–

ErbB4 signaling alters brain functions is whether it also involvesthe E-I imbalance in adulthood. A related question is whetherSZ-associated endophenotypes caused by ErbB4 deficiency inearly development could be diminished by restoring ErbB4 ex-pression in adult animals. These questions require temporalcontrol of ErbB4 mutation and expression. In this paper, wedeveloped genetic approaches; in inducible knockout (iKO)

Significance

NRG1–ErbB4 signaling is implicated in GABAergic circuit as-sembly during development and GABAergic transmission atadulthood. However, it is unclear whether phenotypes in theadult stage in ErbB4 mutant mice result from abnormal neuraldevelopment. By using two strains of mice with temporalcontrol of ErbB4 deletion and expression, we demonstrate thatErbB4 deletion in adult mice impaired behavior and GABA re-lease, whereas deficits due to ErbB4 null mutation during de-velopment were alleviated by restoring ErbB4 expression atthe adult stage. Together, our results indicate that NRG1–ErbB4signaling at adulthood is critical to GABAergic transmission andbehavior and suggest that restoring NRG1–ErbB4 signaling atthe postdevelopmental stage might benefit relevant braindisorders.

Author contributions: H.W., F.L., W. Chen, B.-M.L., W.-C.X., and L.M. designed research; H.W.,F.L., W. Chen, X.S., W. Cui, Z.D., K.Z., H.Z., H.L., G.X., E.F., and B.-X.P. performed research; H.W.,F.L., and W. Chen analyzed data; and H.W., F.L., W. Chen, and L.M. wrote the paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

Published under the PNAS license.1H.W., F.L., and W. Chen contributed equally to this work.2To whom correspondence should be addressed. Email: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1811287115/-/DCSupplemental.

Published online November 29, 2018.

www.pnas.org/cgi/doi/10.1073/pnas.1811287115 PNAS | December 18, 2018 | vol. 115 | no. 51 | 13105–13110

NEU

ROSC

IENCE

Dow

nloa

ded

by g

uest

on

Dec

embe

r 6,

202

0

Page 2: Genetic recovery of ErbB4 in adulthood partially restores ... · Genetic recovery of ErbB4 in adulthood partially restores brain functions in null mice Hongsheng Wanga,1, Fang Liub,1,

mice, ErbB4 expression is normal until tamoxifen (Tam) treat-ment, whereas in recovery knockout (rKO) mice, ErbB4 is ab-sent during development but can be restored in adulthood uponTam treatment. We studied behaviors and characterizedGABAergic transmission of Tam-treated iKO and rKO mice.ErbB4 deletion in adult mice was sufficient to cause behavioraland synaptic deficits; on the other hand, behavioral and synapticdeficits observed in ErbB4 null mice could be diminished byrestoring ErbB4 expression at the adult stage. These resultsprovide compelling evidence that ErbB4 is critical for synaptictransmission and plasticity after development and suggest thatrestoring ErbB4 signaling could be beneficial to relevant SZ.

ResultsBehavioral Deficits in Mice Lacking ErbB4 in Adulthood. To deleteErbB4 at the adult stage, we generated iKO mice by crossingErbB4f/f mice with CAG::Cre-ER mice (Fig. 1A). CAG::Cre-ERmice express a fusion protein consisting of Cre recombinase and amodified ligand-binding domain of the estrogen receptor (ER) un-der the control of the ubiquitous promoter CAG (CMV enhancer,chicken β-actin promoter, rabbit β-globin polyA). In ErbB4f/f mice,exon 2 is floxed, and exon 2 deletion generates a frame shift todisrupt ErbB4 expression. In the resulting CAG::Cre-ER;ErbB4f/f

(iKO) mice, expression of ErbB4 continues to be controlled by thepromoter of the endogenous ErbB4 gene. iKO mice were injectedwith Tam, whose metabolite binds to ER and activates the Cre.Therefore, ErbB4 is expressed in iKO mice until Tam injection. Weinjected iKO mice with Tam or vehicle (referred to as iKO+Tamand iKO+Veh mice) at 8 wk of age and analyzed ErbB4 expression6 wk after (Fig. 1B). As shown in Fig. 1 C and D, ErbB4 expressionin the cortex and hippocampus was abolished in iKO+Tam micecompared with samples from iKO+Veh mice that were similar towild-type mice. Thus, ErbB4 was expressed at a normal level in iKOmice during development but was ablated upon Tam induction.Next, we determined whether adult ErbB4 deletion alters mousebehavior and focused on the paradigms where ErbB4 null mice werefound to be deficient (Fig. 1 E–M). As shown in Fig. 1 E–M, iKO+Veh mice behaved similarly to WTmice in all paradigms, suggestingno apparent effect of the Cre transgene. In contrast, compared withiKO+Veh mice, iKO+Tam mice were hyperactive in an open-field

test as travel distance was increased (Fig. 1 E–G). They were alsoimpaired in prepulse inhibition (PPI) (Fig. 1 H and I) and socialinteraction (Fig. 1 J and K) as PPI ratio was lower while time spentwith stranger mice was reduced. In addition, freezing time wasreduced in iKO+Tam mice in contextual fear conditioning com-pared with iKO+Veh mice (Fig. 1 L and M). Notice that thefreezing times among groups during training were similar (Fig.1M), suggesting a normal ability to sense or escape from footshock. These results indicate that ErbB4 in adult mice is necessaryfor proper behavior. Interestingly, impairment in iKO+Tam micein open-field (Fig. 1 E–G) and PPI tests (Fig. 1 H and I) was lesssevere than in ErbB4 null mice, but that in social interaction (Fig.1 J and K) and contextual memory (Fig. 1 L and M) was similarbetween the two genotypes. This suggests that the ErbB4 nullmutation was more damaging than adult deletion and ErbB4 inadults is more critical for selective behavioral paradigms.

Decreased Inhibitory Transmission in iKO+Tam Mice. Morphologicalstudies of iKO+Tam mice indicate that adult ErbB4 deletion hadlittle effect on global structures and numbers of interneuronspyramidal neurons of the cortex and hippocampus (SI Appendix,Fig. S1). ErbB4 mutation in excitatory neurons has no effect ondendrites of pyramidal neurons (47). In agreement, iKO+Tammice displayed a similar dendrite length and complexity of CA1pyramidal neurons of the hippocampus (SI Appendix, Fig. S2).ErbB4 null and interneuron-specific mutation caused deficits inspines (3, 47, 55, 56) and excitatory synapses onto interneurons(3, 4, 8). To determine whether similar changes may result frompostdevelopmental ErbB4 deletion, we first crossed iKO micewith Thy1-GFP mice to label spines. Quantification of GFP-labeled spines in the hippocampus CA1 region showed spinedensity and morphology in iKO+Tam mice similar to those iniKO+Veh mice and ErbB4f/f mice (SI Appendix, Fig. S3 A–E). Inagreement, there was no change in miniature excitatory post-synaptic current (mEPSC) frequency or amplitude of CA1 py-ramidal neurons (SI Appendix, Fig. S3 F–I). These resultsdemonstrate that adult ErbB4 has little effect on excitatorysynapses between pyramidal neurons or their function. ErbB4 ispresent at postsynaptic sites of excitatory synapses onto in-hibitory neurons and is implicated in their formation (3, 4, 8).

G

Trav

el d

ista

nce

(cm

) H Pulse

PulsePrepulse

response

Stim

ulus

inte

nsity

Time

a

b0

5000

10000

15000 #****

$$

DWTErbB4 nullErbB4f/f

iKO+VehiKO+Tam

CT HCRel

ativ

e in

tens

ity(E

rbB

4/β-

actin

)

0.00.5

1.0

1.5** **

$$ $$** **

ErbB4f/f ErbB4 null iKO+Veh iKO+TamE F

5 10 15 20 25 300500

1000150020002500

$$

**

***

$$#

**

#** $$

** ** **

* $

ErbB4f/f

ErbB4 nulliKO+VehiKO+Tam

Trav

el d

ista

nce

(cm

)

Postnatal weeks0 2 4 6 8 10

Tamoxifen

12 14

Analysis

16 18

B CA

xNormal ErbB4

ErbB4 KO

Veh

TamErbB4f/f

CAG::Cre-ERTM

CAG::Cre-ERTM;ErbB4f/f (iKO)

iKO+Veh

iKO+Tam

ErbB4β-actin

ErbB4β-actin

CT

HC

L

24 h

K

Tim

e in

cha

mbe

rs(s

) M

Free

zing

(%tim

e) ErbB4f/f

iKO+VehiKO+Tam

ErbB4 null

S1 Middle S2050

100150200250

* * *$

*$

020

40

60

80 *$$**

Training Test

I

*** **

** ** **##$$ **

##$$**

$

$PP

I (%

)

70 75 80 85 db020406080

100 J

Tim

e in

cham

bers

(s)

S1 Middle Object050

100150200250

** ***$$ $

ErbB4f/f

iKO+VehiKO+Tam

ErbB4 null

Startle

min

Fig. 1. Behavioral deficits in Tam-treated iKO mice. (A) Breeding diagram of iKO mice. (B) Times of Tam injection and analysis. Tam was injected once everyother day for 20 d. (C and D) Diminished ErbB4 expression in iKO+Tam mice and ErbB4 null mice. From left to right, WT, ErbB4 null, ErbB4f/f, iKO+Veh, andiKO+Tam; n = 3 mice in each group. CT, cortex; HC, hippocampus. (E) Representative travel traces of mice in an open-field test. (F) Increased travel distance(per 5 min) by iKO+Tam mice. (G) Increased total travel distance (within 30 min) by iKO+Tam mice. (H) Diagram of the PPI test. PPI (%) = 100 × (a − b)/a. (I)Impaired PPI of iKO+Tam mice. (J) Decreased preference for the social chamber by iKO+Tam mice. (K) Decreased preference for social novelty by iKO+Tammice. (L) Diagram of contextual fear memory test. (M) Unaltered freezing response to foot shock in training session and reduced freezing time in test sessionby iKO+Tam mice. n = 12 per group for behavior tests; *P < 0.05, **P < 0.01, compared with ErbB4f/f mice; #P < 0.05, ##P < 0.01, compared with ErbB4 nullmice; $P < 0.05, $$P < 0.01, compared with iKO+Veh mice.

13106 | www.pnas.org/cgi/doi/10.1073/pnas.1811287115 Wang et al.

Dow

nloa

ded

by g

uest

on

Dec

embe

r 6,

202

0

Page 3: Genetic recovery of ErbB4 in adulthood partially restores ... · Genetic recovery of ErbB4 in adulthood partially restores brain functions in null mice Hongsheng Wanga,1, Fang Liub,1,

We investigated whether these synapses are altered by adultErbB4 deletion by using Vglut1 and PV antibodies to label, re-spectively, presynaptic excitatory terminals and postsynapticdendrites of PV+ interneurons, many of which are ErbB4+ (4,13, 15, 16). As shown in SI Appendix, Fig. S3 J and K, thenumbers of Vglut1+ puncta onto PV+ dendrites were similaramong different genotypes, regardless of Tam treatment. Inagreement, there was no difference in mEPSC frequency oramplitude of interneurons in the stratum oriens and stratumpyramidale of the hippocampus CA1 region (SI Appendix, Fig. S3L–O). These results indicate no detectable effect of adult ErbB4deletion on excitatory synapses onto interneurons.Studies of ErbB4 null or PV-specific mutation demonstrated

that ErbB4 is necessary for GABAergic transmission in thehippocampus and cortex (3, 4, 17–24, 55). We next determinedwhether this function requires ErbB4 in adulthood by recordingIPSCs (inhibitory postsynaptic currents) in CA1 pyramidal neu-rons (Fig. 2A). As shown in Fig. 2B, evoked IPSC (eIPSC) am-plitude was reduced in iKO+Tam slices, indicating a necessaryrole of adult ErbB4 in maintaining GABA transmission in thehippocampus. This reduction may be caused by a reduction inGABA release and/or GABA receptor density on postsynapticmembrane. To address this question, we compared miniatureIPSC (mIPSC) frequency and amplitude between iKO+Tam andiKO+Veh mice. There was no change in mIPSC amplitude,suggesting GABA receptor density was not compromised (Fig.2D). However, mIPSC frequency was reduced (Fig. 2E), whichmay suggest fewer numbers of inhibitory synapses or diminishedrelease probability. Next, we quantified the numbers of PV-labeled perisomatic inhibitory synapses onto CA1 pyramidalneurons, which were similar among different genotypes, re-gardless of Tam treatment (Fig. 2 F and G), suggesting that re-duced mIPSC frequency may be due to a problem with releaseprobability. To test this hypothesis, we characterized the paired-pulse ratios (PPRs) by recording eIPSCs in response to tandemstimuli with different intervals. The PPRs were increased iniKO+Tam mice compared with ErbB4f/f and iKO+Veh mice(Fig. 2 H and I), indicating compromised GABA releaseprobability. Besides perisomatic synapses, ErbB4+ neuronsalso form inhibitory synapses onto axon initial segments (AISs) ofpyramidal neurons (3, 4, 16). Adult deletion seemed to have noeffect on the number of these synapses (Fig. 2 J–L). Taken to-gether, these observations suggest that ErbB4 in adult animals isnecessary for GABA activity by maintaining release probability.

Diminished GABA Transmission Deficits by Restoring ErbB4 Expressionin Adult Animals. Next, we determined whether ErbB4 in adult-hood is sufficient for GABA transmission by restoring ErbB4expression in ErbB4 null mice. We generated rKO mice byinserting a loxP-NeotpA-loxP cassette into the first intron (be-tween exons 1 and 2) of the ErbB4 gene to first produce Stop-ErbB4 mice (Fig. 3A). The cassette contained the anti-neomycingene (for embryonic stem cell selection) and a poly-A transcrip-tion stop signal. Homozygous Stop-ErbB4 mice did not expressErbB4 in any tissues or cells and died prematurely due to cardiacdeficits, like ErbB4 null mice. To prevent embryonic lethality,hemizygous Stop-ErbB4 mice were crossed with α-MHC::ErbB4mice, which express ErbB4 specifically in developing heart cells.Finally, Stop-ErbB4;α-MHC::ErbB4 mice were crossed with theaforementioned CAG::Cre-ER mice to generate Stop-ErbB4;α-MHC::ErbB4;CAG::Cre-ER (rKO) mice (Fig. 3B). Therefore,rKO mice do not express ErbB4 in the brain or any other tissueexcept the heart (Fig. 3C). After Tam treatment (Fig. 3 A and B)to release the loxP-NeotpA-loxP cassette, rKO miceexpressed ErbB4 in the brain (Fig. 3 D and E). Notice that al-though ErbB4 recovery is mediated by CAG-CreER, the pro-moter activity of the endogenous ErbB4 gene controls the cells inwhich ErbB4 is expressed. As shown in Fig. 3E, the ErbB4 level inthe brain of rKO mice was restored by Tam treatment to a levelthat was comparable to that of control mice (CAG::Cre-ERmice).ErbB4 null mutation reduces the number of PV+ interneurons

in the cortex (5, 7) and hippocampus (54). In agreement, vehicle-treated rKO mice (that did not express ErbB4, Fig. 3E) displayedfewer PV+ neurons in the cortex (SI Appendix, Fig. S4 A–C) andhippocampus (SI Appendix, Fig. S4 D and E). The reducednumber of PV+ interneurons remained unchanged after Tamtreatment (SI Appendix, Fig. S4 B–E), indicating that interneu-ron migration deficit was not rescued by restoring ErbB4 ex-pression in adult animals. The result is not unexpected becauseinterneuron migration was completed before Tam treatment.GABA transmission in the cortex and hippocampus is reduced

in ErbB4 mutant mice (3, 4, 17–24, 55). We next determinedwhether GABA release deficits due to ErbB4 mutation could berescued by restoring ErbB4 expression in adult animals. Asshown in Fig. 4B, eIPSC amplitude was reduced in hippocampalslices of rKO+Veh mice, in agreement with previous reports (18,19, 21, 25). Remarkably, the reduction was mitigated in rKO+Tam slices (Fig. 4B), indicating that restoring ErbB4 is able toenhance GABAergic transmission. mIPSC frequency was also

0 500 1000 1500 2000

0.3

0.6

0.9

1.2

0

Cum

ulat

ive

prob

abili

ty

0246810

mIP

SC

fre

quen

cy(H

z) *$

Inter-pulse interval (ms)

E

LK iKO+TamErbB4f/f iKO+VehGAD67AnkGMerge GAD67 AnkGMerge GAD67AnkGMerge

ErbB4f/f

iKO+VehiKO+Tam

Cum

ulat

ive

prob

abili

ty

010203040

mIP

SC

am

plitu

de(p

A)

0 40 80 120 160

0.3

0.6

0.9

1.2

0Amplitude (pA)

CBAErbB4f/f

iKO+Veh

iKO+Tam

Basket cell

Pyramidal neuron

Nor

mal

ized

eIP

SC

ampl

itude

(% C

ontro

l)

D*

$

0

50

100

150F

iKO+TamErbB4f/f iKO+Veh

PV

/Neu

NP

V/N

euN

JChandelier cell

Pyramidal neuron

IG

02468

10mμ 01/snotuob +V

P

HErbB4f/f

iKO+Veh

iKO+Tam

**$$

0.20.40.60.81.01.21.4

0 5002501005025Pai

red-

puls

e ra

tio

Inter-pulse interval (ms) 05

10

15

20

GA

D67

+bo

uton

s/1

0 μm

Fig. 2. Decreased inhibitory transmission in Tam-treated iKO mice. (A) Recording diagram. (B) Decreased eIPSC amplitude in iKO+Tam mice. n = 7/8 neuronsfrom three mice. (C) Representative traces of mIPSCs. (Scale bar: 20 pA/s.) (D) No change in mIPSC amplitude. (E) Decreased mIPSC frequency. n = 8/9 neuronsfrom three mice. (F) Representative images of PV+ inhibitory synapses onto NeuN+ postsynaptic somata. (Scale bar: Upper, 5 μm; Lower, 2 μm.) (G) Quan-tification data of F. n = 40/42/42 somata of six mice. (H) Representative IPSC traces induced by paired-pulse stimulation with a 100-ms interval. (Scale bar:250 pA/50 ms.) (I) Increased paired-pulse ratios in iKO+Tam mice. n = 10 neurons from four mice. (J) Diagram of inhibitory synapses onto axon initial segments.(K) Representative images of GAD67+ inhibitory synapses onto AnkG+ axon initial segments. AnkG, Ankyrin G. (Scale bar: 5 μm.) (L) Quantification data of K.n = 42/40/43 neurons of six mice. *P < 0.05, **P < 0.01, compared with ErbB4f/f mice; $P < 0.05, $$P < 0.01, compared with iKO+Veh mice.

Wang et al. PNAS | December 18, 2018 | vol. 115 | no. 51 | 13107

NEU

ROSC

IENCE

Dow

nloa

ded

by g

uest

on

Dec

embe

r 6,

202

0

Page 4: Genetic recovery of ErbB4 in adulthood partially restores ... · Genetic recovery of ErbB4 in adulthood partially restores brain functions in null mice Hongsheng Wanga,1, Fang Liub,1,

reduced in rKO+Veh slices (Fig. 4 C–E), in agreement withprevious studies (3, 4, 21, 23). Remarkably, after Tam treatment,the reduction was diminished (i.e., in rKO+Tam slices) (Fig. 4C–E), indicating a rescue effect by adult ErbB4 expression. Todetermine whether this effect resulted from increased numbersof inhibitory synapses, we quantified the number of perisomaticinhibitory synapses onto CA1 pyramidal neurons. As shown inFig. 4 F and G, there was no difference between Tam- and Veh-treated rKO mice, suggesting that the rescue effect on mIPSCfrequency may be mediated by improved release probability. Thisnotion was supported by recovered PPRs (Fig. 4 H and I). Aswith perisomatic synapses, adult recovery of ErbB4 had littleeffect on the number of inhibitory synapses onto AIS of pyra-midal neurons, which remained low in Tam-treated rKO mice(Fig. 4 J–L). mIPSC amplitude was not changed regardless ofgenotypes and treatment (Fig. 4E). Taken together, these ob-servations suggest that restoring ErbB4 in adult animals is able torescue functional deficits of GABA transmission without in-creasing the number of interneurons or inhibitory synapses.

No Effect on Excitatory Synaptic Deficits by Adulthood ErbB4Expression. As stated above, ErbB4 mutation has no effect ondendrites of pyramidal neurons in vivo (47); in agreement, rKO+Veh mice showed a similar dendrite number and complexity (SIAppendix, Fig. S5). To determine whether ErbB4 expression mayrescue deficits of excitatory synapses, we examined their mor-phological and functional properties in Veh- and Tam-treatedrKO mice. As shown in SI Appendix, Fig. S6 A–E, spines of CA1pyramidal neurons, in particular mushroom-shaped spines, were

reduced in Veh-treated rKO mice (that did not express ErbB4), inagreement with previous reports (3, 47, 55, 56), suggesting thatErbB4 is required for spine development. However, the spinedeficits were not diminished by ErbB4 recovery (i.e., in rKO+Tammice). mEPSC frequency of CA1 pyramidal neurons was reducedin rKO+Veh mice compared with control (SI Appendix, Fig. S6 F–I), in agreement with reduced spine number. There was no dif-ference between mEPSC frequencies of Veh- and Tam-treatedrKO mice (SI Appendix, Fig. S6 F–I), indicating that adult ErbB4expression was unable to rescue deficits of excitatory synapses ontopyramidal neurons. The number of excitatory synapses onto in-terneurons is reduced after early ErbB4 mutation (3, 4, 8), asobserved in rKO+Veh mice (SI Appendix, Fig. S6 J and K). Thiswas associated with a reduction in mEPSC frequency of inter-neurons (SI Appendix, Fig. S6 L–O). Both of these deficits (re-duced number of excitatory synapses and mEPSC frequency)remained in rKO+Tam mice (SI Appendix, Fig. S6 J–O), indicatingthat they could not be rescued by adult ErbB4 expression.

Mitigated Behavioral Deficits by Restoring ErbB4 Expression in AdultMice. To determine whether adult ErbB4 expression mitigatesbehavioral deficits caused by ErbB4 null mutation during de-velopment, we characterized behaviors of Veh- and Tam-treatedrKO mice. Compared with control mice, rKO+Veh mice werehyperactive in an open field as travel distance was increased (Fig.5 A–C). They were also impaired in PPI (Fig. 5 D and E) andcontextual fear conditioning (Fig. 5 F and G). Remarkably, thesebehavioral deficits were ameliorated by Tam treatment in rKOmice. Compared with Veh-treated rKO mice, Tam-treated rKO

A

NeotpAF

R

loxp loxpexon1 exon2Stop-ErbB4

rKO-ErbB4+Tam

loxpexon1 exon2

WTexon1 exon2 B

xStop-ErbB4

α-MHC::ErbB4

xCAG::Cre-ERTM

Stop-ErbB4;α-MHC::ErbB4

Veh

Tam

KO in all cellsexcept heart

Recoveredin all cells

rKO

rKO+Veh

rKO+Tam

CErbB4β-actin

D ControlErbB4 nullrKO+VehrKO+Tam

Rel

ativ

e in

tens

ity(E

rbB

4/β-

actin

)E

ErbB4

GAPDHErbB4β-actin

CT

HC0.0

0.5

1.0

1.5

****

$$

****

$$

CT HP

Fig. 3. Generation and characterization of rKOmice. (A) ErbB4 alleles of WT, Stop-ErbB4, and rKO+Tam mice. (B) Breeding diagram of rKO mice. (C )Expression of ErbB4 in Stop-ErbB4;α-MHC::ErbB4mice. (D and E ) Restored ErbB4 expression in thecortex and hippocampus of rKO+Tam mice. CT, cor-tex; HC, hippocampus; from left to right: control,ErbB4 null, iKO+Veh double, iKO+Tam. n = 4 mice;**P < 0.01, compared with control, $$P < 0.01, com-pared with rKO+Veh mice.

A DCControl

rKO+Veh

rKO+Tam

EBasket cell

Pyramidal neuron

0 500 1000 1500 2000Inter-event interval (ms)

0.3

0.6

0.9

1.2

0

Cum

ulat

ive

prob

abili

ty

mIP

SC

fre

quen

cy (H

z)

02468

10

0 40 80 120 160Amplitude (pA)

0.3

0.6

0.9

1.2

0

Cum

ulat

ive

prob

abili

ty

010203040

mIP

SC

am

plitu

de (p

A)

**$

Nor

mal

ized

eIP

SC

ampl

itude

(%C

ontro

l)

B

0

50

100

150

**$

ControlrKO+VehrKO+Tam

KControl rKO+Veh rKO+TamGAD67 AnkGMerge GAD67 AnkGMerge GAD67 AnkGMerge

LJChandelier cell

Pyramidal neuron

G I

PV

+mμ 01/snotuob

02468

10**

HControl

rKO+Veh

rKO+Tam0.20.40.60.81.01.21.4

0 5002501005025Pai

red-

puls

e ra

tio

Inter-pulse interval (ms)

**$$

F

PV

/Neu

NP

V/N

euN

Control rKO+Veh rKO+Tam

05

10

15

20

GA

D67

+bo

uton

s

*

/10

μm

Fig. 4. Diminished GABA transmission deficits in Tam-treated rKO mice. (A) Recording diagram. (B) Diminished reduction of eIPSC amplitude in rKO+Tam mice.n = 7/8 neurons from three mice. (C) Representative traces of mIPSCs. (Scale bar: 20 pA/s.) (D) Diminished reduction of mIPSC frequency in rKO+Tam mice. (E) Nodifference in mIPSC amplitude among groups. n = 8/9 neurons from three mice. (F) Representative images of PV+ inhibitory synapses onto NeuN+ postsynapticsomata. (Scale bar:Upper, 5 μm; Lower, 2 μm.) (G) No effect of Tam treatment on decreased number of inhibitory synapses. n = 45/45/47 somata of seven mice. (H)Representative IPSC traces induced by paired-pulse stimulation with a 100-ms interval. (Scale bar: 250 pA/50 ms.) (I) Recovered paired-pulse ratios in Tam-treatedrKO mice. n = 9/10 neurons of four mice. (J) Diagram of inhibitory synapses onto axon initial segments. (K) Representative images of GAD67+ inhibitory synapsesonto AnkG+ postsynaptic axon initial segments. AnkG, ankyrin G. (Scale bar: 5 μm.) (L) No effect of Tam treatment on decreased number of inhibitory synapses.n = 45/44/47 neurons of seven mice. *P < 0.05, **P < 0.01, compared with control, $P < 0.05, $$P < 0.01, compared with rKO+Veh mice.

13108 | www.pnas.org/cgi/doi/10.1073/pnas.1811287115 Wang et al.

Dow

nloa

ded

by g

uest

on

Dec

embe

r 6,

202

0

Page 5: Genetic recovery of ErbB4 in adulthood partially restores ... · Genetic recovery of ErbB4 in adulthood partially restores brain functions in null mice Hongsheng Wanga,1, Fang Liub,1,

mice were less hyperactive in open field (Fig. 5 A–C), displayedbetter PPI (Fig. 5 D and E), and showed increased freezing timein contextual fear conditioning (Fig. 5 F and G), indicating im-proved behavioral scores by restoring ErbB4 in adult mice. No-tice that the recovery from behavioral deficits was incomplete;that is, Tam-treated rKO mice remained deficient in these par-adigms compared with control mice. These results suggest thatrestoring ErbB4 expression in adulthood mitigates but does noteliminate behavioral deficits caused by ErbB4 loss of functionduring development.

DiscussionThis study determined the role of ErbB4 in the adult brain byutilizing mouse lines that enable temporal control of ErbB4deletion or expression. We showed that adult deletion of ErbB4caused behavioral deficits and reduced IPSCs in hippocam-pal slices, indicating compromised GABAergic transmission.However, it had no effect on numbers of interneurons, inhibitorysynapses onto excitatory neurons, and excitatory synapses in thehippocampus. These results suggest that ErbB4 is critical toGABAergic transmission in adult mice. On the other hand, rKOmice did not express ErbB4 during development and displayedbehavioral deficits similar to those of ErbB4 null mutant mice,including hyperactivity in an open field and impaired PPI andcontextual fear memory. Remarkably, these deficits were di-minished after Tam treatment, indicating that deficits caused byErbB4 deletion during development could be mitigated by re-storing ErbB4 expression at the adult stage. Notice that numbersof PV+ interneurons and inhibitory and excitatory synapsesremained depressed after Tam treatment, suggesting that re-storing ErbB4 in adult animals was unable to diminish structuraldeficits caused by ErbB4 loss of function during development.Together, these observations support a working hypothesis thatrestoring ErbB4 at adulthood could improve GABAergic trans-mission even on compromised circuits and thus diminish behav-ioral deficits that are caused by developmental ErbB4 mutation.ErbB4 is expressed in developing and mature interneurons (4,

13–17) and in mature interneurons, is present in somata, den-drites, and, arguably, axons. ErbB4 is necessary for interneuronmigration from ganglion eminences to the cerebral cortex andforming synapses onto and from pyramidal neurons (25, 26, 57).In the adult cortex and hippocampus, ErbB4 is almost exclusivelyexpressed in GAD67+ interneurons, not in pyramidal neurons(4, 13–16), and has been implicated in maintaining GABAergictransmission and synaptic plasticity (3, 4, 17–24, 55). ErbB4 nullmutant mice displayed fewer numbers of PV+ interneurons (5, 7,54) and compromised GABA release (17, 19, 20, 23). Adultablation of ErbB4 impaired GABAergic transmission, in furthersupport of the hypothesis that GABAergic transmission requiresErbB4. However, the number of PV+ neurons was similar iniKO and control mice, suggesting that adult ErbB4 is not re-quired for the migration and survival of PV+ interneurons withinthe experimental period. Similarly, restoring ErbB4 expressionin adults improved GABAergic transmission compared with thatin ErbB4 null mice but was unable to increase the number of PV+

interneurons (because interneuron migration had already finished).Molecular mechanisms by which ErbB4 regulates interneuron migra-tion, synapse formation onto pyramidal neurons and interneurons, andGABA releases are complex. For example, maintaining GABArelease requires kinase activity, whereas interneuron migration andsynapse formation could also be mediated by kinase-independent, cell-adhesion-dependent mechanisms (6, 58). Future studies are warrantedto determine which mechanism is involved in mitigating deficitscaused by ErbB4 null mutation. Evidently, ErbB4 level or promoteractivity is dominantly expressed in interneurons (4, 13–17). However,ErbB4 knockdown was shown to reduce spines in vitro (56), andpyramidal neuron-specific deletion of ErbB4 could reduce thenumber of spines of pyramidal neurons in vivo (47, 59). These resultssuggest a role of ErbB4 in pyramidal neurons. Our data are unableto determine whether phenotypes were due to loss of ErbB4 in in-terneurons and/or pyramidal neurons. For example, loss of spinescould be due to the loss of ErbB4 in pyramidal neurons or a com-pensatory mechanism due to ErbB4 loss in interneurons (3, 55, 60).Earlier studies suggested NRG1–ErbB4 signaling is involved

in controlling E-I balance by maintaining GABAergic trans-mission. First, NRG1 increases mIPSCs and eIPSCs in corticaland hippocampal slices (17, 18, 20, 24). This is likely due to in-creased GABA release probability (17, 23, 24). Second, this effectrequires ErbB4 because it is diminished by pharmacological in-hibition of the kinase activity of ErbB4 or genetic deletion of theErbB4 gene in null or interneuron-specific mutant mice (17, 18,21, 24). Third, treating brain slices with an NRG1-neutralizingpeptide decreases mIPSCs and eIPSCs (17, 18, 20, 21, 23, 24,61). Fourth, NRG1 neutralization and/or ErbB4 mutationimpair behaviors in various paradigms involving prefrontal cor-tex, the hippocampus, and the amygdala (3, 15, 18–24), whereErbB4 is enriched. Results from iKO and rKO mice add to thenotion that ErbB4 is critical to E-I balance in the adult brain.In summary, our study demonstrates a role of ErbB4 signaling at

the adult stage in maintaining normal GABAergic transmission, E-Ibalance, and behaviors. The results suggest that abnormal ErbB4function in adult animals could contribute to the pathophysiologicalmechanism of relevant disorders. A mouse model mimicking highNRG1 levels in patients (32, 62) displayed SZ-related deficits (46).Such deficits could be rescued by reducing the NRG1 level at theadult stage (46). Together, these observations suggest that relevantpatients could benefit from therapeutic approaches of optimizingNRG1–ErbB4 signaling.

Materials and MethodsAnimals, electrophysiology, immunofluorescence, Western blot, Golgistaining, analysis of spine morphology, behavioral tests, and statisticalanalyses are described in SI Appendix. Experimental procedures were ap-proved by the Institutional Animal Care and Use Committees of AugustaUniversity and Case Western Reserve University.

ACKNOWLEDGMENTS. We thank Dr. Cary Lai for ErbB4 antibodies and Dr.Martin Gassmann for MHC-ErbB4 transgenic mice. This work is supported byNIH Grants MH083317, MH109280, NS082007, and NS090083 (to L.M.) andAG051773 and AG045781 (to W.-C.X.).

C

D EP

PI (

%)

Pulse

PulsePrepulse

response

Stim

ulus

inte

nsity

Time

a

b

ArKO+Veh rKO +TamControl

B

Trav

el d

ista

nce

(cm

)

0

5000

10000

15000 ** $

Trav

el d

ista

nce

(cm

) *

F G

Free

zing

(% ti

me)

24 h

**$$

*

70 75 80 85 db0

20

40

60

80

**$ **

**$*

ControlrKO+VehrKO+Tam

5 10 15 20 25 300

1000

2000

3000 ** ** ** ****$

$ $$ $$* * *

0

20

40

6080

Training Test

Startle

min

Fig. 5. Mitigated behavioral deficits in Tam-treatedrKO mice. (A) Representative travel traces of mice inan open-field test. (B) Mitigation of increased traveldistance (per 5 min) in rKO+Tam mice. (C) Mitigationof increased total travel distance (within 30 min) inrKO+Tam mice. (D) Diagram of the PPI test. PPI (%) =100 × (a − b)/a. (E ) Mitigation of impaired PPI inrKO+Tam mice. (F ) Diagram of contextual fearmemory test. (G) Mitigation of reduced freezing timein rKO+Tam mice. n = 12 mice; *P < 0.05, **P < 0.01,compared with control; $P < 0.05, $$P < 0.01, com-pared with rKO+Veh mice.

Wang et al. PNAS | December 18, 2018 | vol. 115 | no. 51 | 13109

NEU

ROSC

IENCE

Dow

nloa

ded

by g

uest

on

Dec

embe

r 6,

202

0

Page 6: Genetic recovery of ErbB4 in adulthood partially restores ... · Genetic recovery of ErbB4 in adulthood partially restores brain functions in null mice Hongsheng Wanga,1, Fang Liub,1,

1. McGrath J, Saha S, Chant D, Welham J (2008) Schizophrenia: A concise overview ofincidence, prevalence, and mortality. Epidemiol Rev 30:67–76.

2. Cahill ME, et al. (2012) Control of interneuron dendritic growth through NRG1/erbB4-mediated kalirin-7 disinhibition. Mol Psychiatry 17:99–107.

3. Del Pino I, et al. (2013) Erbb4 deletion from fast-spiking interneurons causes schizo-phrenia-like phenotypes. Neuron 79:1152–1168.

4. Fazzari P, et al. (2010) Control of cortical GABA circuitry development by Nrg1 andErbB4 signalling. Nature 464:1376–1380.

5. Flames N, et al. (2004) Short- and long-range attraction of cortical GABAergic inter-neurons by neuregulin-1. Neuron 44:251–261.

6. Krivosheya D, et al. (2008) ErbB4-neuregulin signaling modulates synapse develop-ment and dendritic arborization through distinct mechanisms. J Biol Chem 283:32944–32956.

7. Li H, Chou SJ, Hamasaki T, Perez-Garcia CG, O’Leary DDM (2012) Neuregulin repellentsignaling via ErbB4 restricts GABAergic interneurons to migratory paths from gan-glionic eminence to cortical destinations. Neural Dev 7:10.

8. Ting AK, et al. (2011) Neuregulin 1 promotes excitatory synapse development andfunction in GABAergic interneurons. J Neurosci 31:15–25.

9. Yang JM, et al. (2013) Development of GABA circuitry of fast-spiking basket inter-neurons in the medial prefrontal cortex of erbb4-mutant mice. J Neurosci 33:19724–19733.

10. Bartolini G, et al. (2017) Neuregulin 3 mediates cortical plate invasion and laminarallocation of GABAergic interneurons. Cell Rep 18:1157–1170.

11. Liu X, et al. (2011) Specific regulation of NRG1 isoform expression by neuronal ac-tivity. J Neurosci 31:8491–8501.

12. Ozaki M, Itoh K, Miyakawa Y, Kishida H, Hashikawa T (2004) Protein processing andreleases of neuregulin-1 are regulated in an activity-dependent manner. J Neurochem91:176–188.

13. Vullhorst D, et al. (2009) Selective expression of ErbB4 in interneurons, but not py-ramidal cells, of the rodent hippocampus. J Neurosci 29:12255–12264.

14. Neddens J, et al. (2011) Conserved interneuron-specific ErbB4 expression in frontalcortex of rodents, monkeys, and humans: Implications for schizophrenia. BiolPsychiatry 70:636–645.

15. Bean JC, et al. (2014) Genetic labeling reveals novel cellular targets of schizophreniasusceptibility gene: Distribution of GABA and non-GABA ErbB4-positive cells in adultmouse brain. J Neurosci 34:13549–13566.

16. Lin TW, et al. (2018) Regulation of synapse development by Vgat deletion fromErbB4-positive interneurons. J Neurosci 38:2533–2550.

17. Woo RS, et al. (2007) Neuregulin-1 enhances depolarization-induced GABA release.Neuron 54:599–610.

18. Wen L, et al. (2010) Neuregulin 1 regulates pyramidal neuron activity via ErbB4 inparvalbumin-positive interneurons. Proc Natl Acad Sci USA 107:1211–1216.

19. Tan GH, et al. (2011) Neuregulin 1 represses limbic epileptogenesis through ErbB4 inparvalbumin-expressing interneurons. Nat Neurosci 15:258–266.

20. Lu Y, et al. (2014) Maintenance of GABAergic activity by neuregulin 1-ErbB4 inamygdala for fear memory. Neuron 84:835–846.

21. Bi LL, et al. (2015) Amygdala NRG1-ErbB4 is critical for the modulation of anxiety-likebehaviors. Neuropsychopharmacology 40:974–986.

22. Li KX, et al. (2011) Neuregulin 1 regulates excitability of fast-spiking neurons throughKv1.1 and acts in epilepsy. Nat Neurosci 15:267–273.

23. Tan Z, et al. (2018) Dynamic ErbB4 activity in hippocampal-prefrontal synchrony andtop-down attention in rodents. Neuron 98:380–393.e4.

24. Chen YJ, et al. (2010) ErbB4 in parvalbumin-positive interneurons is critical forneuregulin 1 regulation of long-term potentiation. Proc Natl Acad Sci USA 107:21818–21823.

25. Mei L, Nave KA (2014) Neuregulin-ERBB signaling in the nervous system and neuro-psychiatric diseases. Neuron 83:27–49.

26. Mei L, Xiong WC (2008) Neuregulin 1 in neural development, synaptic plasticity andschizophrenia. Nat Rev Neurosci 9:437–452.

27. Schizophrenia Working Group of the Psychiatric Genomics Consortium (2014) Bi-ological insights from 108 schizophrenia-associated genetic loci. Nature 511:421–427.

28. Agim ZS, et al. (2013) Discovery, validation and characterization of Erbb4 and Nrg1haplotypes using data from three genome-wide association studies of schizophrenia.PLoS One 8:e53042.

29. Munafò MR, Attwood AS, Flint J (2008) Neuregulin 1 genotype and schizophrenia.Schizophr Bull 34:9–12.

30. Munafo MR, Thiselton DL, Clark TG, Flint J (2006) Association of the NRG1 geneand schizophrenia: A meta-analysis. Mol Psychiatry 11:539–546, and erratum (2006)11:613.

31. Stefansson H, et al. (2002) Neuregulin 1 and susceptibility to schizophrenia. Am J HumGenet 71:877–892.

32. Weickert CS, Tiwari Y, Schofield PR, Mowry BJ, Fullerton JM (2012) Schizophrenia-associated HapICE haplotype is associated with increased NRG1 type III expression andhigh nucleotide diversity. Transl Psychiatry 2:e104.

33. Li D, Collier DA, He L (2006) Meta-analysis shows strong positive association of the

neuregulin 1 (NRG1) gene with schizophrenia. Hum Mol Genet 15:1995–2002.34. Gong YG, et al. (2009) A two-method meta-analysis of neuregulin 1(NRG1) associa-

tion and heterogeneity in schizophrenia. Schizophr Res 111:109–114.35. Walker RM, et al. (2010) Association analysis of neuregulin 1 candidate regions in

schizophrenia and bipolar disorder. Neurosci Lett 478:9–13.36. Kukshal P, et al. (2013) Association study of neuregulin-1 gene polymorphisms in a

north Indian schizophrenia sample. Schizophr Res 144:24–30.37. Suárez-Pinilla P, et al. (2015) Progressive structural brain changes and NRG1 gene

variants in first-episode nonaffective psychosis. Neuropsychobiology 71:103–111.38. Grimm O, et al. (2014) Striatal response to reward anticipation: Evidence for a sys-

tems-level intermediate phenotype for schizophrenia. JAMA Psychiatry 71:531–539.39. Greenwood TA, et al. (2016) Genetic assessment of additional endophenotypes from

the Consortium on the Genetics of Schizophrenia Family Study. Schizophr Res 170:

30–40.40. Wang D, et al. (2015) Large-scale candidate gene study to identify genetic risk factors

predictive of paliperidone treatment response in patients with schizophrenia.

Pharmacogenet Genomics 25:173–185.41. Jajodia A, et al. (2015) Evaluation of genetic association of neurodevelopment and

neuroimmunological genes with antipsychotic treatment response in schizophrenia in

Indian populations. Mol Genet Genomic Med 4:18–27.42. Kampman O, et al. (2004) Neuregulin genotype and medication response in Finnish

patients with schizophrenia. Neuroreport 15:2517–2520.43. Chung DW, et al. (2016) Dysregulated ErbB4 splicing in schizophrenia: Selective ef-

fects on parvalbumin expression. Am J Psychiatry 173:60–68.44. Wang R, et al. (2015) Decreased plasma levels of neureglin-1 in drug naïve patients

and chronic patients with schizophrenia. Neurosci Lett 606:220–224.45. Marballi K, Cruz D, Thompson P, Walss-Bass C (2012) Differential neuregulin 1

cleavage in the prefrontal cortex and hippocampus in schizophrenia and bipolar

disorder: Preliminary findings. PLoS One 7:e36431.46. Yin DM, et al. (2013) Reversal of behavioral deficits and synaptic dysfunction in mice

overexpressing neuregulin 1. Neuron 78:644–657.47. Barros CS, et al. (2009) Impaired maturation of dendritic spines without disorgani-

zation of cortical cell layers in mice lacking NRG1/ErbB signaling in the central nervous

system. Proc Natl Acad Sci USA 106:4507–4512.48. Chen YJJ, et al. (2008) Type III neuregulin-1 is required for normal sensorimotor

gating, memory-related behaviors, and corticostriatal circuit components. J Neurosci

28:6872–6883.49. Mostaid MS, et al. (2017) Meta-analysis reveals associations between genetic variation

in the 5′ and 3′ regions of neuregulin-1 and schizophrenia. Transl Psychiatry 7:e1004.50. Jagannath V, Gerstenberg M, Correll CU, Walitza S, Grünblatt E (2018) A systematic

meta-analysis of the association of neuregulin 1 (NRG1), D-amino acid oxidase (DAO),

and DAO activator (DAOA)/G72 polymorphisms with schizophrenia. J Neural Transm

(Vienna) 125:89–102.51. Feng Y, et al. (2017) Association between ErbB4 single nucleotide polymorphisms and

susceptibility to schizophrenia: A meta-analysis of case-control studies. Medicine

(Baltimore) 96:e5920.52. Marenco S, et al. (2011) Genetic association of ErbB4 and human cortical GABA levels

in vivo. J Neurosci 31:11628–11632.53. Luykx JJ, et al. (2012) A common variant in ERBB4 regulates GABA concentrations in

human cerebrospinal fluid. Neuropsychopharmacology 37:2088–2092.54. Neddens J, Buonanno A (2010) Selective populations of hippocampal interneurons

express ErbB4 and their number and distribution is altered in ErbB4 knockout mice.

Hippocampus 20:724–744.55. Yin DM, et al. (2013) Regulation of spine formation by ErbB4 in PV-positive inter-

neurons. J Neurosci 33:19295–19303.56. Li B, Woo RS, Mei L, Malinow R (2007) The neuregulin-1 receptor erbB4 controls

glutamatergic synapse maturation and plasticity. Neuron 54:583–597.57. Rico B, Marín O (2011) Neuregulin signaling, cortical circuitry development and

schizophrenia. Curr Opin Genet Dev 21:262–270.58. Müller T, et al. (2018) Neuregulin 3 promotes excitatory synapse formation on hip-

pocampal interneurons. EMBO J, 37:e98858.59. Cooper MA, Koleske AJ (2014) Ablation of ErbB4 from excitatory neurons leads to

reduced dendritic spine density in mouse prefrontal cortex. J Comp Neurol 522:

3351–3362.60. Wang DD, Kriegstein AR (2008) GABA regulates excitatory synapse formation in the

neocortex via NMDA receptor activation. J Neurosci 28:5547–5558.61. Geng F, et al. (2016) Neuregulin 1-ErbB4 signaling in the bed nucleus of the stria

terminalis regulates anxiety-like behavior. Neuroscience 329:182–192.62. Hahn CG, et al. (2006) Altered neuregulin 1-erbB4 signaling contributes to NMDA

receptor hypofunction in schizophrenia. Nat Med 12:824–828.

13110 | www.pnas.org/cgi/doi/10.1073/pnas.1811287115 Wang et al.

Dow

nloa

ded

by g

uest

on

Dec

embe

r 6,

202

0