https://www.cell.com/cell-reports/fulltext/S2211-1247(19)31449-4?_returnURL=https%3A%2F%2Flinkinghub.elsevier.com%2Fretrieve%2Fpii%2FS2211124719314494%3Fshowall%3Dtrue

Comment; Mitogen-activated protein kinase (MAPK) phosphorylates Npas4 in Dopamine-1 receptor-Medium Spiny Neurons within the Nucleus Accumbens increasing transcriptional activity to enhance reward-related learning and memory via protein kinase A (PKA)/Rap1. This is the molecular basis for control of protein formation involved in stimulant abuse/addiction.

Open AccessDOI:https://doi.org/10.1016/j.celrep.2019.10.116PlumX MetricsPrevious ArticleLiver Soluble Epoxide Hydrolase Regulates Behavi …Next ArticleMuscarinic Modulation of Antennal Lobe GABAergic L …

Highlights

  • •More than 400 CBP-interacting proteins, including Npas4, are identified
  • •MAPK phosphorylates Npas4 and increases the interaction between Npas4 and CBP
  • •Phosphorylation of Npas4 enhances the transcriptional activity of Npas4
  • •Phosphorylation of Npas4 in D1R-MSNs regulates reward-related learning and memory

Summary

Dopamine (DA) activates mitogen-activated protein kinase (MAPK) via protein kinase A (PKA)/Rap1 in medium spiny neurons (MSNs) expressing the dopamine D1 receptor (D1R) in the nucleus accumbens (NAc), thereby regulating reward-related behavior. However, how MAPK regulates reward-related learning and memory through gene expression is poorly understood. Here, to identify the relevant transcriptional factors, we perform proteomic analysis using affinity beads coated with cyclic AMP response element binding protein (CREB)-binding protein (CBP), a transcriptional coactivator involved in reward-related behavior. We identify more than 400 CBP-interacting proteins, including Neuronal Per Arnt Sim domain protein 4 (Npas4). We find that MAPK phosphorylates Npas4 downstream of PKA, increasing the Npas4-CBP interaction and the transcriptional activity of Npas4 at the brain-derived neurotrophic factor (BDNF) promoter. The deletion of Npas4 in D1R-expressing MSNs impairs cocaine-induced place preference, which is rescued by Npas4-wild-type (WT), but not by a phospho-deficient Npas4 mutant. These observations suggest that MAPK phosphorylates Npas4 in D1R-MSNs and increases transcriptional activity to enhance reward-related learning and memory.

Graphical Abstract

Figure thumbnail fx1
View Large ImageFigure ViewerDownload Hi-res imageDownload (PPT)

Introduction

Dopamine (DA) is important for motor function, motivation, working memory, and the reward system (Girault and Greengard, 2004Phillips et al., 2008). Functional deficits in DA signaling have been implicated in various neuropsychological diseases, including Parkinson’s disease, drug addiction, compulsive behavior, autism spectrum disorders, and schizophrenia (Carlsson, 2001Hyman et al., 2006Iversen and Iversen, 2007Koob and Volkow, 2010Swanson et al., 2007). Experiencing reward and using various drugs of abuse, such as cocaine or methamphetamine, activate intracellular pathways in the brain reward system, including the ventral tegmental area (VTA), the nucleus accumbens (NAc), and the prefrontal cortex (Rogge and Wood, 2013). These pathways regulate the expression of genes that are essential for long-lasting forms of synaptic plasticity, memory processes, and drug-induced neuronal and behavioral changes (McClung and Nestler, 2008Renthal and Nestler, 2008). Despite this knowledge, our current understanding of the mechanisms by which some transcriptional factors (TFs) are stimulated by DA and how they regulate several genes that are important for synaptic plasticity remains incomplete. It is even less well-understood how the changes that occur at the intracellular level manifest in behavioral changes.The principal target of DA is medium spiny neurons (MSNs), which are a special type of GABAergic inhibitory cell that comprises 90% of the neurons within the striatum, including the NAc. The MSNs in the NAc receive inputs from the dopaminergic neurons of the VTA and the glutamatergic neurons of the hippocampus, amygdala, and medial prefrontal cortex. There are two distinct classes of spatially intermixed MSNs that express dopamine type 1 or 2 receptors (D1R-MSNs or D2R-MSNs, respectively; Smith et al., 2013). D1R-MSN and D2R-MSN subpopulations were also originally characterized as direct and indirect pathways in the dorsal striatum. D1R is coupled to adenylate cyclase through Golf (Gs) and activates protein kinase A (PKA), whereas D2R inhibits adenylate cyclase through Gi (Hervé et al., 1993Stoof and Kebabian, 1984). We previously found that DA stimulates Rap1 GEF (Rasgrp2) phosphorylation via PKA in D1R-MSNs, thereby activating Rap1, and then Rap1 regulates neuronal excitability and behavioral responses to cocaine reward through mitogen-activated protein kinase 1/3 (MAPK1/3), also known as extracellular-signal-regulated kinase 1/2 (ERK1/2) (Nagai et al., 2016). MAPK1/3 is thought to phosphorylate several potassium and HCN channels that are potentially involved in neuronal excitability (Nagai et al., 2016). Alternatively, MAPK1/3 may phosphorylate several nuclear TFs that are involved in gene expression, long-term synaptic plasticity, and memory formation. However, it remains unclear how MAPK1/3 regulates gene expression and memory formation downstream of the D1R/PKA/Rap1 pathway.TFs such as c-Fos, FosB, and cyclic AMP response element (CRE) binding protein (CREB) are activated by DA and mediate the expression of genes that are involved in synaptic plasticity (Hyman and Malenka, 2001Nestler, 2001Robinson and Kolb, 1999). A variety of protein kinases, including PKA, RSK, and CaMKII, activate CREB via serine-133 (S133) phosphorylation (Choe and McGinty, 2000Choe and McGinty, 2001Dash et al., 1991Montminy et al., 1990Sheng et al., 1991Xing et al., 1996Xing et al., 1998). The phosphorylation of CREB at S133 recruits CREB-binding protein (CBP) to promote transcription. CBP and its homolog p300 are essential transcriptional coactivators for many TFs (Karamouzis et al., 2007Vo and Goodman, 2001). CBP/p300 can also act as a scaffold protein, stabilizing the transcription complex by simultaneously binding to other proteins. CBP also regulates transcription during memory and synaptic plasticity (Barrett and Wood, 2008). The deletion of CBP in the NAc correlates with impairments in cocaine sensitivity and reward-related learning and memory (Malvaez et al., 2011Rogge and Wood, 2013). However, how CBP cooperates with many TFs during reward-related memory processing is not fully understood.Here, we isolated and concentrated TFs from the mouse striatum using affinity beads coated with CBP, which acts as a transcriptional coactivator, and identified several TFs, including Neuronal Per Arnt Sim domain protein 4 (Npas4). MAPK phosphorylated Npas4 downstream of PKA. The phosphorylation of Npas4 increased the interaction between Npas4 and CBP, thereby regulating transcriptional activity to enhance reward-related learning and memory.

Results

 Proteomic Analysis of CBP-Binding Proteins

To concentrate and identify TFs that are associated with reward-related learning and memory, we subjected mice to cocaine-induced conditioned place preference (CPP) conditioning and performed a glutathione-S-transferase (GST) pull-down assay followed by shotgun analysis using liquid chromatography-tandem mass spectrometry (LC‐MS/MS). CPP is a behavioral paradigm based on the principles of Pavlovian conditioning (Tzschentke, 2007). In the conditioning session of the CPP, mice learn to associate the rewarding effects of cocaine to a neutral environmental context that has been paired with cocaine administration. We injected cocaine into mice and placed them into the CPP test chamber for 30 min, after which nuclear extracts from the striatal region, including the NAc, were applied to affinity beads coated with GST or the GST-CBP-N-terminal transactivation domain (N-TAD), which can interact with several TFs, such as CREB (Karamouzis et al., 2007Vo and Goodman, 2001Figure 1A). Numerous proteins were eluted from the affinity beads coated with GST‐CBP-N-TAD, whereas fewer proteins from affinity beads coated with GST (Figure 1B). The bound proteins were digested using trypsin/Lys-C and subjected to LC-MS/MS to identify interacting proteins. By the shotgun analysis, we identified more than 400 proteins that specifically interact with GST-CBP-N-TAD, but not with GST (Tables 1 and S1). To confirm the results obtained in the proteomic analysis, we performed an immunoblot analysis using specific antibodies. CREB, Histone H3, FosB, ΔFosB, and Jun interacted with GST-CBP-N-TAD, but not GST, as previously described (Figure 1C; Parker et al., 1996Tsai et al., 2008Vo and Goodman, 2001). CBP-interacting candidates, including Npas4, MKL/myocardin-like protein 2 (Mkl2), Matrin-3, and transcriptional activator protein Pur-alpha (purine-rich element-binding protein alpha), also specifically interacted with GST-CBP-N-TAD, but not GST (Figure 1C). To investigate the functions of the identified CBP-interacting proteins, we performed gene ontology (GO) analysis (biological process) and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis using the DAVID bioinformatics resources (Huang da et al., 2009aHuang da et al., 2009b). We found that most CBP-interacting proteins were classified into RNA and transcription-related proteins, and some were also classified into synaptic plasticity and learning-related proteins in GO analysis (Figure 1D). KEGG pathway analysis showed that the CBP-interacting proteins were involved not only in spliceosome and mRNA surveillance but also in long-term potentiation, amphetamine addiction, and dopaminergic synapse (Figure 1D).

Figure thumbnail gr1
Figure 1Npas4 Is a Novel CBP-Interacting ProteinShow full captionView Large ImageFigure ViewerDownload Hi-res imageDownload (PPT)

Table 1List of Identified CBP-Interacting Proteins

Akap5Akap8Kiaa1210Ablim2Arf3Actn1Actn2Actn4
Add1Apba2Anks1bAnkrd63Ank2Ank3Ap2a1Ap2a2
Ap2b1Ap2m1Amer2Agap2Ahdc1Atp5c1PfkpDhx9
Ddx1Ddx18Ddx3xDdx3yDdx50Dhx36Atad3Aurkaip1
Epb41l3Add2No66Bmp1Baiap2BcanBrd7Brd9
Btbd17Kctd16Mapk8ip3Cdh2Calcoco1Slc25a12Calm1Camsap3
CaluCamkvCsnk1eCsnk2a1ClpbCaap1Ctnna1Ctnna2
Ctnnb1Ctnnd2CtifCdc5lCenpvPde2aCspg5Chtop
Cbx3Cbx6Chd4Chd5CltcCldn11Clvs2Cpsf1
Cpsf2Cpsf3Cpsf4Ccdc136Csde1CirbpC3C1qbp
Cnksr2Fam120bH2afyH2afy2Coro1cCoro2bCkmt1Creb1
Cdk12Cyb561d1Cyth3Cyfip2Cpeb2Cpeb3Specc1Dbp
Bdh1Znf326DgkbDgkzTfb1mDlg2Dlg3Dlgap3
Polr2bPolr2eDnajaDnaja3Dnajc10Adarb1Dynll1Dynll2
Mib2Nedd4Smurf1Egr3Entpd4Elavl1Elavl3Slc4a4
TufmNgefEsrp1Erc2Eif4a3Slc1a2Capza1Capza2
CapzbFbxo3Fbxo41Fbxw10Fez1Farp1Fbln5Fmr1
Fxr1Fxr2Fn3kAldoaFyco1Grsf1Gtf3c1Gtf3c5
Gtf3c5Grin2bGrwd1GlulGlrx2Gnb2l1Gnb4Gnb5
Gar1Dkc1Hspa12aHp1bp3HnrnpabHnrnpa0Hnrnpa1Hnrnpdl
HnrnpdlHnrnpmSyncripHnrnpuHnrnpul1Hnrnpul2HnrnpcHk1
H1f0Hist1h1cHist1h1eHist2h2acHist1h2bfH3f3cHist1h4aRbbp4
Kmt2aHomer1Kpnb1Usp53Ing2Ints3Itgb1bp1Ilf2
Ilf3AqrIqsec1Iqsec2Isca2JupKhdrbs1Khdrbs2
Khdrbs3Kif2aLmnb1Lgi1Lgi2Lgi3Lrrc7Cd47
Ppfia2Ppfia3SsbCcm2Map6d1Map7d1Matr3Grm1
Mbd3Mgst3Map1bMap4Mapre3Map1lc3aMast3Slc25a11
Mtfr1lTimm50Mapk1Mkl1Mkl2Mybbp1aMyef2Mag
MogMlf2Myl12bMyh10Myh9Sbf1CmasNaa30
Nwd2Nckap1Ntn1Ppp1r9bNrxn1NfascNpas4Nhp2l1
NonoNficNfatc4Nup133Nup160Nup85Ncor2Nxf1
ExogYbx1Nop16Nop56Nop58Ddx21NclNup43
Nupl2Ptcd3Ppil1CaskSlc25a3Pi4kaPip5k1aPacs1
Pkp4Plrg1Pabpn1Poldip2Kcna1Kcnq2Wdr33Prpf19
Prpf8Bcas2Ddx17Ddx27Ddx6Ythdc2Dimt1Smarca2
Rbm19Usp9xPhb2Akt1s1Prr3Lrp1Psma8Bsn
Pdia4EmsyFam131bFam98aFam98bFosbPrkcbPrkcg
Lin7cMagohPptc7IkRufy3Soga3Sec31bUnc13a
WizPurgGlyr1Dhx15Rapgef4G3bp2Rap1aPtprs
Rims1Rgs9Rcn2Arhgef2RhogPrps1Rsl1d1Rrs1
Rpf2Rimbp2Rbfox1Rbfox3Rbm10Rbm14Rbm4bRbm7
Rbm8aFusMsi2RalyFblFbll1Ruvbl1Ruvbl2
Skp1SarnpAtp2a2Safb2Sept7StrapSrrm2Mark2
Cdc42bpaCdc42bpbSik3Ppp2r2aPpp2r2dPpp2r2cPpp2r1aPpp6c
Ppp6r3Ppp1caShank3ScocSfxn3SnrpeSnrpfSnrpg
Snrpd1SnrpnSnw1Atp1a1Atp1a3Atp1b1Slc2aSlc25a48
Sorbs2StrbpDdx39bSf3a1Sf3b1Sf3b3Sf3b4Sf3b6
SfpqSrcin1Srpk1Srpk2Hspa9Stip1Strn4Strip2
SdhaSdhbSucla2Sun1Surf6Sugp2Smarcb1Synpo
Snap25Syt7SnphTardbpTnrTns2Zcchc6Spock2
AlyrefThy1TefTjp1Tjp2Smarca4Eny2Jun
PuraPurbCtcfTra2aTgfb1i1Slc25a1Trim3Trim46
TrioRtcbTmod1Tmod2Tmod3Tmod4Tubal3Tinagl1
Baz1bSnrpcSnrpa1Prpf3Snrnp200Snrnp40Fyttd1Usp46
Rps27aGm996Myo5aCn166Ubp1Hacd3Vdac1Vdac2
Vdac3Kcnab2Wdr37Wdr47Wdr6Wdr7Wdr82Smu1
Zbtb7aZc3h11aZc3h4Zcrb1Zmat4Znf148Znf281Znf512
Znf638Znf771ZfrEftud2YwhabYwhaeYwhahYwhag
YwhaqYwhazOgdh

The nuclear extracts of the striatal and NAc were applied to affinity beads coated with GST or GST-CBP-N-TAD. The bound proteins were digested using trypsin/Lys-C and subjected to LC-MS/MS to identify the proteins. To select proteins that specifically interact with CBP, the proteins from the control GST column were removed from the data. High-abundance proteins (such as keratin and ribosomal protein) were also excluded.

Among the CBP-interacting candidates in the categories of synaptic plasticity and learning, further experiments focused on Npas4. Npas4 is a brain-specific basic helix-loop-helix transcriptional factor that is expressed throughout the whole brain at a low level in the resting state, though it is enriched in the limbic system, such as the hippocampus, amygdala, and NAc (Choy et al., 2015Leong et al., 2013Ooe et al., 2004). Npas4 plays a role in the expression of activity-dependent genes, such as c-Fos and brain-derived neurotrophic factor (BDNF) to control synaptic plasticity (Bloodgood et al., 2013Lin et al., 2008Pruunsild et al., 2011Ramamoorthi et al., 2011Spiegel et al., 2014). Npas4 is also required for normal social interaction and contextual memory formation in mice (Coutellier et al., 2012Ramamoorthi et al., 2011). We examined whether CPP conditioning would affect the interaction between CBP and Npas4 and found that the interaction increased after CPP conditioning (Figure 1E). It has been reported that Npas4 was expressed in multiple cell types, including MSNs, in the NAc (Taniguchi et al., 2017). However, it is unknown whether Npas4 is expressed in D1R-MSNs or D2R-MSNs. To provide more accurate information of Npas4 expression in MSNs, we compared the expression of Npas4 in Drd1-mVenus and Drd2-mVenus transgenic mice that were injected with cocaine and placed into the CPP chamber. We found that more than 75% of Npas4-expressing cells were D1R-MSNs positive, and less than 25% of Npas4-expressing cells were D2R-MSNs positive in the NAc (Figure 1F). Npas4 colocalized with CBP in the nuclei (Figure 1F). These results suggest that Npas4 associates with CBP in the nuclei of D1R-MSNs in the NAc under CPP conditioning.

 MAPK Regulates the Interaction between CBP and Npas4

To examine the Npas4-binding region of CBP, we transfected GFP-Npas4-wild-type (WT) into COS7 cells and performed a GST pull-down assay using various GST-CBP fragments as bait. Npas4 mainly bound to the N-TAD (228–683 amino acids [aas]) of CBP. Npas4 weakly bound to the C-TAD (1,891–2,442 aas), but not to the nuclear receptor interaction domain (RID) (1–227 aas), the Bromo (684–1,172 aas), or the histone acetyltransferase (HAT) (1,173–1,890 aas) domains (Figures 2A and 2B ). To examine the CBP-binding region of Npas4, we transfected Myc-CBP-N-TAD into COS7 cells and performed a GST pull-down using various GST-Npas4 fragments as bait. CBP mainly bound to the TAD (598–802 aas) of Npas4. CBP weakly bound to the middle domain (274–597 aas), but not to the basic helix-loop-helix (bHLH)-Per Arnt Sim (PAS) domain (1–273 aas; Figures 2C and 2D). These results indicate that the N-TAD domain of CBP mainly binds to Npas4 274–802 aas.

Figure thumbnail gr2
Figure 2MAPK Regulates the Interaction between CBP and Npas4Show full captionView Large ImageFigure ViewerDownload Hi-res imageDownload (PPT)

It has been reported that the kinase-inducible interaction (KIX) domain of CBP, which is located in the CBP-N-TAD, interacts with the kinase-inducible domain (KID) of CREB and that this interaction is increased by the phosphorylation of CREB at S133 (Chrivia et al., 1993). Thus, we speculated that the interaction between CBP and Npas4 was also regulated by phosphorylation. To examine whether the phosphorylation of Npas4 modulates its interaction with CBP, we performed a GST pull-down assay using okadaic acid (OA), a phosphatase inhibitor, to increase Npas4 phosphorylation. We also used specific inhibitors for several kinases, including MAP2K (MEK), c-jun N-terminal kinase (JNK), p38 MAPK, glycogen synthase kinase-3 (GSK3), PKA, and phosphoinositide 3-kinase (PI3K). These kinase inhibitors specifically inhibited OA-induced phosphorylation of each kinase substrate (Figure S1). The treatment of GFP-Npas4-WT-expressing COS7 cells with OA induced a band shift of GFP-Npas4-WT and increased the interaction between GFP-Npas4-WT and GST-CBP-N-TAD, and this interaction was inhibited by U0126 (MAP2K inhibitor), but not by SP600125 (JNK inhibitor), SB203580 (p38 MAPK inhibitor), SB216763 (GSK3 inhibitor), H89 (PKA inhibitor), and LY294002 (PI3K inhibitor; Figure 2E). We also found that the cotransfection of the constitutively active (CA) form of MAP2K1 into COS7 cells induced a band shift of GFP-Npas4-WT and increased its interaction with GST-CBP-N-TAD, although neither MAP2K1-WT nor the dominant-negative (DN) form of MAP2K1 showed such effects (Figure 2F). These results suggest that MAPK phosphorylates Npas4 and, consequently, increases its interaction with CBP.

 Identification of Sites Where MAPK Phosphorylates Npas4

Next, we examined whether MAPK directly phosphorylates Npas4. An in vitro phosphorylation assay revealed that MAPK1 effectively phosphorylated Npas4 274–597 aas and 598–802 aas, but not 1–273 aas (Figures 3A and 3B ), suggesting that MAPK1 directly phosphorylates Npas4 274–802 aas in vitro. To further investigate whether MAPK phosphorylates Npas4 in COS7 cells, we performed Phos-tag SDS-PAGE, in which phosphorylated protein has a slower migration rate on the gel as compared to nonphosphorylated proteins (Kinoshita et al., 2006). GFP-Npas4-WT was prominently shifted upward when GFP-Npas4-WT was coexpressed with MAP2K1-CA, but not with MAP2K1-WT or DN (Figure 3C), indicating that Npas4 is phosphorylated by MAPK in COS7 cells. To determine which region of Npas4 is phosphorylated by MAPK in COS7 cells, we divided Npas4 274–802 aas into five fragments: Npas4 274–389 aas; 390–489 aas; 490–597 aas; 598–701 aas; and 702–802 aas (Figure 3A). Npas4 390–489 aas, 490–597 aas, and 598–701 aas appeared as several bands (Figure 3D), and these bands were strongly shifted upward in the presence of MAP2K1-CA. In contrast, neither Npas4 274–389 aas nor Npas4 702–802 aas showed any upward shift when coexpressed with MAP2K1-CA (Figure 3D). These results indicate that MAPK phosphorylation sites exist in Npas4 390–701 aas. To identify the phosphorylation sites of Npas4, pulled down GST-Npas4 fragments coexpressed with MAP2K1-CA from COS7 cells were digested with trypsin/Lys-C at the C terminus of lysine and arginine residues or Glu-C/Asp-N at the C terminus of glutamic acid residues and N terminus of aspartic acid residues and then peptides were analyzed by LC-MS/MS. Dual-phosphorylated peptide DLVCTPPYTPHQPGGCAFLFSLHEPFQTHLPPPSSSLQE, containing T423 and T427 phosphorylation sites, was identified from digested fragments of GST-Npas4-390–489 aas; LPPSPSSPGNGDCTLLALAQLR, containing S577 and S580 phosphorylation sites, was identified from digested fragments of GST-Npas4 490–597 aas; and GLLTPEASPVKQSFFHYTEKE, containing T611 and S615 phosphorylation sites, was identified from digested fragments of GST-Npas4 598–701 aas (Figure S2). Selected ion monitoring (SIM) analysis revealed that the amount of this dual phosphorylation was significantly increased by coexpression with MAP2K1-CA (Figure 3E). To confirm the phosphorylation sites identified by mass spectrometry, we constructed alanine (Ala) mutants of all the (S/T)P sites in the Npas4 fragments and expressed them in COS7 cells with or without coexpression with MAP2K1-CA. The band shifts of Npas4 390–489 aas induced by coexpression with MAP2K1-CA were lost in the T423A/T427A mutant (Figure 3F). The band shifts of Npas4 490–597 aas induced by coexpression with MAP2K1-CA were lost in the S577A/S580A mutant (Figure 3G). The top band shift of Npas4 598–701 aas among the three bands induced by coexpression with MAP2K1-CA was lost in the T611A/S615A mutant (Figure 3H). To confirm the phosphorylation of these sites in full length of Npas4-WT, we mutated T423, T427, S577, S580, T611, and S615 to Ala, producing what we designated the Npas4-6A mutant. The MAPK-dependent band shift of Npas4 was decreased in the Npas4-6A mutant (Figure 3I). Taken together, these results indicate that Npas4 is phosphorylated by MAPK at T423, T427, S577, S580, T611, and S615 sites.

Figure thumbnail gr3
Figure 3Identification of Sites of MAPK-Dependent Npas4 PhosphorylationShow full captionView Large ImageFigure ViewerDownload Hi-res imageDownload (PPT)

 Phosphorylation of Npas4 by MAPK in Striatal Neurons

MAPK is a proline-directed kinase, meaning that it phosphorylates a consensus motif of serine-proline (SP) or threonine-proline (TP). Among the phosphorylation sites of Npas4, T427, S577, and S580 completely match the consensus motif P-X-S/T-P (where X represents any amino acid); T423, T611, and S615 match the consensus motif S/T-P. T423 and T427 are located near the putative docking site for MAPK, known as the DEF domain (docking site for ERK and FXF) (Figure 4A). These phosphorylation sites are well conserved in mammals (human, mouse, and rat; Figure 4A). To monitor the phosphorylation state of Npas4 by MAPK, we prepared antibodies that specifically recognized Npas4 phosphorylated at T427, S577, S580, and S615 (anti-pT427, anti-pS577, anti-pS580, and anti-pS615 antibodies), but we failed to prepare antibodies to recognize Npas4 phosphorylated at T423 and T611. The specificity of these antibodies was examined with immunoblot analysis. GST-Npas4 274–597 aas or GST-Npas4 598–802 aas phosphorylated by MAPK in vitro was specifically detected by the anti-pT427, anti-pS577, anti-pS580, and pS615 antibodies in a dose-dependent manner (Figure S3). We also confirmed that the anti-pT427, anti-pS577, anti-pS580, and pS615 antibodies did not cross-react with nonphosphorylated Npas4 (Figure S3).

Figure thumbnail gr4
Figure 4Phosphorylation of Npas4 by MAPKShow full captionView Large ImageFigure ViewerDownload Hi-res imageDownload (PPT)

To examine whether MAPK phosphorylates Npas4 at T427, S577, S580, and S615 in COS7 cells, GFP-Npas4-WT was transfected into COS7 cells with or without the coexpression of MAP2K1-CA. Coexpression with MAP2K1-CA induced the phosphorylation of Npas4 at T427, S577, S580, and S615 (Figure 4B). We confirmed that the anti-pT427, anti-pS577, anti-pS580, and anti-pS615 antibodies did not cross-react with the Npas4-6A mutant and the Npas4 point mutant (Figures 4B and S4). Because the pT427 antibody is the most sensitive, in the following experiment, the pT427 antibody was used to monitor intracellular phosphorylation. The treatment of COS7 with OA induced the phosphorylation of not only MAPK1/3 but also Npas4 at T427 (Figure 4C). The pretreatment of the cells with U0126 inhibited the OA-induced phosphorylation of Npas4 at T427 (Figures 4C and S1), whereas the pretreatment with JNK, p38 MAPK, GSK3, PKA, and PI3K inhibitors did not inhibit the OA-induced phosphorylation of Npas4 at T427 in COS7 cells (Figure S1). These results suggest that MAPK specifically phosphorylates Npas4 at T427 in COS7 cells. To explore Npas4 phosphorylation under more physiologically relevant conditions, we used cultured striatal neurons. It has been reported that Npas4 protein expression can be induced in cultured neurons by depolarization (50 mM KCl treatment; Lin et al., 2008). Because Npas4 expression levels are very low under normal conditions, it is difficult to detect the endogenous phosphorylation of Npas4. Thus, we pretreated cells with KCl for 1 h to increase Npas4 expression. It has been reported that activation of MAPK by depolarization peaks at 5 min and returns to steady state after 1 to 2 h (Tyssowski et al., 2018). The stimulation of striatal neurons with OA induced the phosphorylation of not only MAPK1/3 but also endogenous Npas4 at T427 (Figure 4D). The pretreatment of the cells with U0126 inhibited the OA-induced phosphorylation of Npas4 (Figure 4D). We previously reported that the treatment of striatal neurons with forskolin (FSK), which specifically produces cyclic AMP (cAMP) and subsequent activation of PKA, increases Rap1 GEF (Rasgrp2) phosphorylation via PKA, and the phosphorylation of Rasgrp2 leads to Rap1 activation, followed by recruitment of the MAPK pathway (Nagai et al., 2016). The stimulation of striatal neurons with FSK induced the phosphorylation of not only Rasgrp2 and MAPK1/3 but also endogenous Npas4 at T427 (Figure 4E). The pretreatment of the cells with U0126 inhibited the FSK-induced phosphorylation of both MAPK1/3 and Npas4, but not Rasgrp2. These results suggest that MAPK phosphorylates Npas4 downstream of the cAMP/PKA/Rap1 pathway (Figure 4E).Next, we investigated the spatial distribution of phosphorylated Npas4. Unfortunately, the anti-pT427 antibody could not detect the phosphorylation of endogenous Npas4 during immunostaining of the striatal neurons. Accordingly, GFP-Npas4-WT was transfected into cultured striatal neurons to visualize Npas4 and its phosphorylated form. GFP-Npas4-WT was abundantly distributed in the nucleus and also distributed in the cell body (Figure 4F). The stimulation of striatal neurons with FSK induced the phosphorylation of GFP-Npas4-WT at T427 in the nuclei (Figure 4F). Taken together, these results suggest that Npas4 is phosphorylated by MAPK downstream of the cAMP/PKA/Rap1 pathway in the nuclei of cultured striatal neurons.

 The Effects of Npas4 Phosphorylation on Its Interaction with CBP; Npas4 Phosphorylation Regulates Transcriptional Activity

To examine whether the MAPK-mediated phosphorylation of Npas4 affects its interaction with CBP, we performed a GST pull-down assay using COS7 cells expressing GFP-Npas4-WT or Npas4-6A. The stimulation of the COS7 cells with OA increased the interaction between GST-CBP-N-TAD and GFP-Npas4-WT, but not with Npas4-6A (Figure 5A). To mimic the phosphorylation of Npas4 by MAPK, we substituted glutamic acid for T423, T427, S577, S580, T611, and S615 to produce GFP-Npas4-6E. GST-CBP-N-TAD interacted with GFP-Npas4-6E more than with GFP-Npas4-WT (Figure 5B). These results suggest that the phosphorylation of Npas4 by MAPK increases its interaction with CBP in COS7 cells.

Figure thumbnail gr5
Figure 5Effects of the Phosphorylation of Npas4 on Its Interaction with CBP; Transcriptional Regulation by Npas4 PhosphorylationShow full captionView Large ImageFigure ViewerDownload Hi-res imageDownload (PPT)

To measure the transcriptional activity of Npas4, we performed one-hybrid analysis using a luciferase reporter driven by the Gal4-responsive element (Gal4-UAS) and Gal4 DNA-binding domain fused with Npas4 274–802 aas (Gal4-DBD-Npas4 274–802 aas). The transcriptional activity of Npas4 274–802 aas was significantly enhanced by MAP2K1-CA coexpression, as well as FSK treatment (Figure 5C), suggesting that the Npas4 activity was increased downstream of PKA/Rap1/MAPK pathway. It has been reported that Npas4 regulates the expression of several genes, such as Bdnf, that are important for neuronal survival and synaptic plasticity (Lin et al., 2008Pruunsild et al., 2011Spiegel et al., 2014Yun et al., 2013). BDNF plays a crucial role in synaptic plasticity and memory processing in the adult brain (Bekinschtein et al., 2008). The levels of Bdnf mRNA are increased in the NAc following acute and repeated cocaine treatment (Filip et al., 2006). To examine whether the MAPK-mediated phosphorylation of Npas4 modulates BDNF promoter activity, we performed a dual-reporter assay using the exon 1 BDNF promoter and found that the expression of GFP-Npas4-WT increased exon 1 BDNF promoter activity. This activity was further increased by MAP2K1-CA coexpression and by FSK treatment in cultured striatal neurons (Figures 5D and 5E). The expression of MAP2K1-CA or treatment of FSK did not significantly increase BDNF promoter activity without the expression of GFP-Npas4-WT (Figures 5D and 5E). We also found that, compared with Npas4-WT, Npas4-6A had reduced ability to activate exon 1 BDNF promoter and Npas4-6E enhanced exon 1 BDNF promoter activity (Figure 5F). These results suggest that the phosphorylation of Npas4 by MAPK increases BDNF promoter activity.

 Npas4 and Its Phosphorylation in Accumbal D1R-MSNs Control Reward-Related Learning and Memory

To examine whether Npas4 is associated with reward-related learning and memory, we performed a cocaine-induced CPP test using Npas4 knockout (KO) mice. Before the CPP test, we checked whether Npas4 KO itself affects the D1R signaling pathway. Peritoneal injection of cocaine induced the phosphorylation of Rasgrp2 and MAPK1/3 in both WT and Npas4 KO mice (Figure S5A), indicating that the cocaine-induced activation of PKA and MAPK was normal in Npas4 KO mice. Cocaine injection induced place preference for the drug-paired side in WT mice, whereas the saline treatment did not result in place preference. The global knockout of Npas4 impaired cocaine-induced place preference (Figure S5B). This result is consistent with a recent study, which found that Npas4 knockout in the NAc impaired CPP (Taniguchi et al., 2017).To further investigate whether Npas4 is required for reward learning and memory in D1R-MSNs or D2R-MSNs, we used Drd1a-Cre transgenic (tg) mice that express Cre-recombinase (Cre) specifically in D1R-MSNs (Gong et al., 2003) or adenosine A2a receptor (Adora2a)-Cre tg mice that express Cre specifically in D2R-MSNs (Gong et al., 2007). We established a system in which the dominant-negative form (DN) of Npas4 (Pruunsild et al., 2011Yoshihara et al., 2014) was expressed in the NAc under the control of the D1R promoter or Adora2a promoter using adeno-associated virus (AAV)-mediated conditional transgene techniques (Figure 6A). The expression in the presence of Cre is achieved by Flex recombination of a transgene that changes the orientation of the coding sequence with respect to the promoter from the anti-sense to sense (Betley and Sternson, 2011Figure 6A). We injected AAV-Flex-mCherry or AAV-Flex-mCherry-P2A-Npas4-DN into the NAc of Drd1a-Cre mice or Adora2a-Cre mice and performed the CPP test 2 to 3 weeks after viral injection (Figure 6B). Cocaine injection induced place preference to the drug-paired side in mCherry transfected mice, whereas the saline did not result in place preference. The rewarding effects of cocaine were markedly reduced in mice that expressed Npas4-DN in D1R-MSNs, but not in D2R-MSNs, in the NAc (Figure 6C). We also employed AAV-substance P (SP)-Cre and homozygous loxP-flanked (floxed) Npas4 mice (Npas4 fl/fl) to examine the specific functions of Npas4 in D1R-MSNs (Figure 6D). AAV-Flex-mCherry and AAV-SP-EGFP were coinjected with or without AAV-SP-Cre into the NAc of Npas4 fl/fl mice, and the CPP test was performed 2 to 3 weeks after viral injection (Figure 6B). The localized knockout of Npas4 in D1R-MSNs markedly decreased cocaine-induced place preference compared with the control AAV injection (Figure 6E). These results indicate that Npas4 in D1R-MSNs is required for reward-related learning and memory. To examine whether the phosphorylation of Npas4 regulates reward-related learning and memory, we coinjected AAV-Flex-mCherry-P2A-Npas4-WT or AAV-Flex-mCherry-P2A-Npas4-6A and AAV-SP-Cre into the NAc of Npas4 fl/fl mice (Figure 6E). The cocaine-induced CPP deficits in mice with local Npas4 knockout were restored by the expression of Npas4-WT, but not Npas4-6A (Figure 6E), suggesting that Npas4 phosphorylation in D1R-MSNs is important for reward-related learning and memory. Furthermore, to examine whether Npas4-6E rescued CPP when MAPK signaling was blocked, we coinfected AAV-SP-Cre and AAV-Flex-EGFP-P2A-MAP2K1-DN and/or AAV-Flex-mCherry-P2A-Npas4-WT or AAV-Flex-mCherry-P2A-Npas4-6E into the NAc (Figures S5C and S5D). The expression of MAP2K1-DN in D1R-MSNs significantly impaired cocaine-induced place preference. This defect could not be completely recovered by coexpression of Npas4-WT or 6E (Figures S5C and S5D), suggesting that additional MAPK substrates are also involved in reward-related learning and memory.

Figure thumbnail gr6
Figure 6Npas4 and Its Phosphorylation in Accumbal D1R-MSNs Control Reward-Related Learning and MemoryShow full captionView Large ImageFigure ViewerDownload Hi-res imageDownload (PPT)

Discussion

 The Functional Significance of Npas4 Phosphorylation by MAPK

There is substantial evidence that Npas4 expression is regulated by activity-dependent Ca2+ signaling (Choy et al., 2015Lin et al., 2008Ramamoorthi et al., 2011) and PI3K-Akt signaling (Ooe et al., 2009Speckmann et al., 2016). However, it is not well understood how Npas4 activity is regulated at the posttranslational level. In the present study, we found that MAPK directly phosphorylated Npas4 in an in vitro kinase assay (Figure 3B) and that MAP2K1-CA induced a clear band shift of Npas4 in Phos-tag SDS-PAGE (Figure 3C). These observations, together with the previous finding that MAP2K inhibitor treatment prevents a band shift of Npas4 in Laemmli’s SDS-PAGE (Ooe et al., 2009Speckmann et al., 2016), indicate that MAPK directly phosphorylates Npas4. Furthermore, with LC-MS/MS and phos-tag SDS-PAGE analysis, we identified the sites of Npas4 phosphorylation by MAPK as T423, T427, S577, S580, T611, and S615 (Figures 3D–3H). These multiple phosphorylation sites are located in the middle region and C-terminal region of the TAD domain, not in the N-terminal region of the DNA-binding domain, suggesting that the MAPK-dependent phosphorylation of Npas4 affects transcriptional activity through protein interaction, but not DNA binding. We found that Npas4 274–802 aas interacted with the N-TAD domain of CBP, which was increased in a MAPK activity-dependent manner (Figures 2E and 2F). Consistently, MAPK signal activation through the expression of MAP2K1-CA or FSK treatment enhanced the transcriptional activity of Npas4 (Figures 5C–5E). Furthermore, the Npas4-6A mutant showed reduced transcriptional activity and the Npas4-6E mutant showed enhanced transcriptional activity (Figure 5F). Taken together, these observations suggest that the phosphorylation of Npas4 by MAPK enhances the transcriptional activity of Npas4 through CBP binding.

 Role of Npas4 in Reward-Related Learning and Memory in D1R-MSNs

It has been reported that Npas4 in the hippocampal CA3 region is required for normal social interaction and contextual fear memory formation (Coutellier et al., 2012Ramamoorthi et al., 2011). However, the functional role of Npas4 in the NAc and reward-related behaviors is not well understood. A recent study has shown that Npas4 knockout in the NAc impairs CPP (Taniguchi et al., 2017). However, virus-based approaches using nonspecific Cre inactivate all neurons in the injection area, and it remains unclear which neuronal population is responsible for Npas4-dependent reward effects. Indeed, we previously found that the selective expression of MAP2K1-CA in D1R-MSNs significantly potentiated methamphetamine-induced conditioned place preference, whereas the expression of MAP2K1-CA in D2R-MSNs in the NAc did not (Bin Saifullah et al., 2018). In the present study, we found that Npas4 was mainly expressed in D1R-MSNs after CPP training (Figure 1F). To analyze D1R-specific Npas4 function, Npas4-DN was expressed in the NAc under the control of the D1R or Adora2a promoter using AAV-mediated conditional transgenic techniques (Figure 6A). The rewarding effects of cocaine were markedly potentiated in mice that expressed Npas4-DN in D1R-MSNs, but not in D2R-MSNs, of the NAc (Figure 6C). We also administered AAV-SP-Cre into the NAc of homozygous floxed Npas4 mice (Npas4 fl/fl) (Figure 6D). The D1R-specific knockout of Npas4 notably diminished cocaine-induced CPP (Figure 6E). This deficit in mice with local knockout of Npas4 was restored by the expression of Npas4-WT, but not Npas4-6A (Figure 6E), indicating that Npas4 phosphorylation in D1R-MSNs is important for reward-related learning and memory.

 Target Genes of Npas4 in Reward-Related Learning and Memory

In the present study, we found that the expression of Npas4 increased exon 1 BDNF promoter activity and that the phosphorylation of Npas4 enhanced its activity (Figures 5D–5F). Npas4 and its phosphorylation were required for cocaine-induced CPP (Figure 6C and 6E). Our findings raise the possibility that Npas4 and its phosphorylation regulate BDNF expression to form long-term reward-related memory. It has been reported that the expression of Bdnf is upregulated in the NAc of animals conditioned in a CPP task (Tian et al., 2016) and that the expression of lentivirus-BDNF and TrkB enhanced cocaine-induced CPP (Bahi et al., 2008). In addition to BDNF, Npas4 can regulate the expression of several genes that are important for neuronal survival and synaptic plasticity, such as c-FosZif268Arc/Arg3.1, and Homer1a (Lin et al., 2008Pruunsild et al., 2011Shan et al., 2018Spiegel et al., 2014Yoshihara et al., 2014). It has been reported that cocaine injection predominantly induces the expression of c-Fos and Zif268 in D1R-expressing MSNs (Bertran-Gonzalez et al., 2008) and increases Arc/Arg3.1 mRNA levels in the striatum (Caffino et al., 2011). Cocaine injection has been shown to induce Homer1a protein expression in the striatum, and this increase was blocked by a D1R antagonist, but not a D2R antagonist (Patil et al., 2007). A recent study has shown that Homer1a deletion reduces cocaine-induced behavioral sensitization but has no effect on cocaine-induced CPP (Datko et al., 2017). Exploring the role of Npas4 target genes in reward-related learning and memory will be an important future direction.

 Roles of CBP-Interacting Proteins

CBP is a multipurpose transcriptional coactivator that participates in a diverse array of basic cellular functions, including DNA repair, cell growth, differentiation, apoptosis, synaptic plasticity, and memory. These functions of CBP are regulated by protein-protein interactions between CBP and other transcriptional regulators. Such interactions regulate gene activation and subsequent cell functions. In this study, a proteomic screening of mouse striatal tissue using the N-terminal transactivation domain of CBP as bait identified several CBP-interacting partners, including CREB, Histone H3, FosB, ΔFosB, Jun, Npas4, Mkl2, Matrin-3, and Pur-alpha (Figures 1A–1C). CREB and ΔFosB are well-known CBP-interacting proteins that are implicated in reward-related behavior (Carlezon et al., 1998Kelz et al., 1999Nestler et al., 1999). Here, we focused specifically on Npas4 and revealed that MAPK regulated the CBP-Npas4 interaction to enhance reward-related memory. Mkl2 and matrin-3 were identified as other potential CBP-interacting partners. Mkl1 (MAL/MRTF-A/BSAC) and Mkl2 (MRTF-B/MAL16) are coactivators that interact with serum response factor (SRF) and regulate SRF-dependent gene expression (Kalita et al., 2012). Mkl1 and Mkl2 are involved in the reorganization of dendritic spines and the complexity of dendrites (Ishikawa et al., 2010). The deletion of SRF from the NAc decreases animals’ sensitivity to the rewarding effects of cocaine (Vialou et al., 2012). It is possible that the interactions among Mkl2, SRF, and CBP cooperate to regulate reward-related memory formation. Matrin-3 is an inner nuclear matrix protein with two zinc finger domains and two RNA recognition motifs (RRMs) (Salton et al., 2011). The nuclear matrix participates in DNA replication, transcription, RNA processing, and chromatin organization. Matrin-3 is reported to be phosphorylated by PKA following the activation of NMDA receptors, a process that leads to its degradation (Giordano et al., 2005). This PKA-mediated phosphorylation of Matrin-3 and the modulation of nuclear matrix function could be involved in long-term potentiation (Giordano et al., 2005). Future research on these CBP-interacting proteins could identify novel mechanisms involved in reward-related learning and memory.

STAR★Methods

 Key Resources Table

REAGENT or RESOURCESOURCEIDENTIFIER
Antibodies
Rabbit monoclonal anti-phospho-CREB (S133)Cell Signaling TechnologyCat# 9198; RRID: AB_2561044
Rabbit monoclonal anti-CREBCell Signaling TechnologyCat# 9197; RRID: AB_331277
Mouse monoclonal anti-CREBCell Signaling TechnologyCat# 9104; RRID: AB_490881
Rabbit monoclonal anti-Histone H3Cell Signaling TechnologyCat# 4499; RRID: AB_10544537
Rabbit monoclonal anti-FosBCell Signaling TechnologyCat# 2251; RRID: AB_2106903
Rabbit monoclonal anti-phospho-p44/42 MAPK (ERK1/2) (T202/Y204)Cell Signaling TechnologyCat# 4370; RRID: AB_2315112
Rabbit monoclonal anti-p44/42 MAPK (ERK1/2)Cell Signaling TechnologyCat# 4695; RRID: AB_390779
Mouse monoclonal anti-p44/42 MAPK (ERK1/2)Cell Signaling TechnologyCat# 9107; RRID: AB_10695739
Rabbit monoclonal anti-phospho-SAPK/JNK (T183 / Y185)Cell Signaling TechnologyCat# 4668; RRID: AB_823588
Rabbit polyclonal anti-SAPK/JNKCell Signaling TechnologyCat# 9252; RRID: AB_2250373
Rabbit monoclonal anti-phospho-p38 MAPK (T180/Y182)Cell Signaling TechnologyCat# 4631; RRID: AB_331765
Mouse monoclonal p38 MAPKCell Signaling TechnologyCat# 9217; RRID: AB_331298
Rabbit monoclonal anti-phospho-AKT (T308)Cell Signaling TechnologyCat# 2965; RRID: AB_2255933
Rabbit monoclonal anti-AKTCell Signaling TechnologyCat# 4691; RRID: AB_915783
Rabbit polyclonal anti-Npas4MerckCat# HPA039255; RRID: AB_10674282
Mouse monoclonal anti-CBP/p300MerckCat# P2859; RRID: AB_260994
Rabbit polyclonal anti-Rasgrp2Thermo Fisher ScientificCat# PA5-35111; RRID: AB_2552421
Rabbit polyclonal anti-Mkl2Novus BiologicalsCat# NBP1-46209; RRID: AB_10009441
Mouse monoclonal anti-JunBD BiosciencesCat# 610327; RRID: AB_397717
Mouse monoclonal anti-CRMP-2IBL-AmericaCat# JP11096; RRID: AB_1630812
Rabbit polyclonal anti-Matrin-3Bethyl LaboratoriesCat# A300-591A; RRID: AB_495514
Rabbit polyclonal anti-Pur-alphaAbcamCat# ab79936; RRID: AB_2253242
Rabbit polyclonal anti-CBP (A-22)Santa Cruz BiotechnologyCat# sc-369; RRID: AB_631006
Rabbit polyclonal anti-c-Myc (A-14)Santa Cruz BiotechnologyCat# sc-789; RRID: AB_631274
Rabbit polyclonal anti-RFPMBL InternationalCat# PM005; RRID: AB_591279
Rabbit polyclonal anti-GFPMBL InternationalCat# 598; RRID: AB_591819
Mouse monoclonal anti-GFPRocheCat# 11814460001; RRID: AB_390913
Rat monoclonal anti-GFPNacalai TesqueCat# GF090R; RRID: AB_2314545
Mouse monoclonal anti-GSTFUJIFILM WakoCat# 017-21854
Rabbit polyclonal anti-phospho-CRMP2 (T514)Yoshimura et al., 2005N/A
Rabbit polyclonal anti-phospho-Rasgrp2 (S116/S117)Nagai et al., 2016N/A
Rabbit polyclonal anti-Npas4Yun et al., 2010N/A
Rabbit polyclonal anti-phospho-Npas4 (T427)This paperN/A
Rabbit polyclonal anti-phospho-Npas4 (S577)This paperN/A
Rabbit polyclonal anti-phospho-Npas4 (S580)This paperN/A
Rabbit polyclonal anti-phospho-Npas4 (S615)This paperN/A
Donkey anti-Rat IgG, Alexa Fluor 488Thermo Fisher ScientificCat# A-21208; RRID: AB_2535794
Donkey anti-Rabbit IgG, Alexa Fluor 555Thermo Fisher ScientificCat# A-31572; RRID: AB_162543
Donkey anti-Mouse IgG, Alexa Fluor 647Thermo Fisher ScientificCat# A-31571; RRID: AB_162542
Goat anti-rabbit IgG, Alexa Fluor 680Thermo Fisher ScientificCat# A-21109; RRID: AB_2535758
Goat anti-Mouse IgG, Alexa Fluor 680Thermo Fisher ScientificCat# A-21058; RRID: AB_2535724
anti-mouse IgG (DyLight 800 Conjugate)Cell Signaling TechnologyCat# 5257; RRID: AB_10693543
IRDye 800CW Goat anti-Mouse IgGLI-COR BiosciencesCat# 925-32210; RRID: AB_2687825
Bacterial and Virus Strains
pAAV-Sp-CreHikida et al., 2010Nagai et al., 2016N/A
pAAV-Sp-EGFPHikida et al., 2010Nagai et al., 2016N/A
pAAV-CAGGS-Flex-EGFP-P2A-MCS-WPRENagai et al., 2016N/A
pAAV-CAGGS-Flex-EGFP-P2A-dnMAP2K1-WPRENagai et al., 2016N/A
pAAV-CaMKII-Flex-mCherry-P2A-MCS-WPREThis paperN/A
pAAV-CaMKII-Flex-mCherry-P2A-Npas4-WT-WPREThis paperN/A
pAAV-CaMKII-Flex-mCherry-P2A-Npas4-6A-WPREThis paperN/A
pAAV-CaMKII-Flex-mCherry-P2A-Npas4-6E-WPREThis paperN/A
pAAV-CaMKII-Flex-mCherry-P2A-Npas4-DN-WPREThis paperN/A
Chemicals, Peptides, and Recombinant Proteins
Okadaic acidLC LaboratoriesCat# O-5857
Forskolin [7β-Acetoxy-8,13-epoxy-1α,6β,9α-trihydroxy-labd-14-en-11-one]MerckCat# 344270
U0126 [1,4-diamino-2,3-dicyano-1,4-bis(o-aminophenylmercapto) butadiene]MerckCat# 662005
SP600125 [Anthra[1,9-cd]pyrazol-6(2H)-one,1,9-pyrazoloanthrone]MerckCat# S5567
H-89 Dihydrochloride N-[2-((p-Bromocinnamyl) amino)ethyl]-5-isoquinolinesulfonamide, 2HCl,MerckCat# 371962
LY294002 [2-(4-Morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one]MerckCat# 440202
SB203580 [4-(4-Fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole]Tocris BioscienceCat# 1202/1
SB216763 [3-(2,4-Dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)-1H-pyrrole-2,5-dione]Tocris BioscienceCat# 1616
cOmplete, Mini, EDTA-free Protease Inhibitor CocktailRocheCat# 11836170001
PhosSTOPRocheCat# 04906837001
Lipofectamine 2000Thermo Fisher ScientificCat# 11668500
Dulbecco’s Modified Eagle’s Medium (DMEM)MerckCat# D5796
Fetal Bovine Serum (FBS)MerckCat# 173012
Neurobasal mediumThermo Fisher ScientificCat# 21103049
B-27 supplementThermo Fisher ScientificCat# 17504044
GlutaMax SupplementThermo Fisher ScientificCat# 35050061
Poly-D-lysine hydrobromideMerckCat# P6407
Neuron Dissociation SolutionsFUJIFILM WakoCat# 297-78101
Glutathione Sepharose 4BGE HealthcareCat# 217075605
Trypsin/Lys-C MixPromegaCat# V5072
Glu-CPromegaCat# V1651
Asp-NFUJIFILM WakoCat# 056-05921
2,2,2-Tribromoethanol (Avertin)FUJIFILM WakoCat# 203-14602
Tert-amylalcohol (Avertin)FUJIFILM WakoCat# 010-03703
active MAPK1Carna BiosciencesCat# 204-143
GST-CBP-RIDThis paperN/A
GST-CBP-N-TADThis paperN/A
GST-CBP-bromoThis paperN/A
GST-CBP-HATThis paperN/A
GST-CBP-C-TADThis paperN/A
GST-Npas4 1-273 aaThis paperN/A
GST-Npas4 274-597 aaThis paperN/A
GST-Npas4 598-802 aaThis paperN/A
Critical Commercial Assays
Dual-Luciferase Reporter Assay SystemPromegaCat# E1910
Nuclear Extract KitActive MotifCat# 40010
PrimeSTAR Mutagenesis Basal KitTakaraCat# R046A
Experimental Models: Cell Lines
COS7 cell lineATCCRRID: CVCL_0224
AAV293 cell lineCell BioLabsRRID: CVCL_KA64
Cultured striatal neurons (from embryos of ICR mice)Cowan et al., 2005N/A
Experimental Models: Organisms/Strains
C57BL/6Japan SLCRRID: IMSR_JAX:000664
ICRJapan SLCRRID: IMSR_JAX:009122
Npas4 KO (Npas4tm1Meg/Npas4tm1Meg)Lin et al., 2008RRID: MGI:3828101
Npas4 flox (Npas4tm2Meg/Npas4tm2Meg)Lin et al., 2008RRID: MGI:3828102
Drd1-mVenus (C57BL/6J-Tg (mDrd1-YFP) 680-1Koba/KobaRbrc)Nagai et al., 2016RRID: IMSR_RBRC03111
Drd2-mVenus (C57BL/6J-Tg (Drd2-YFP) 364-5Koba/KobaRbrc)Nagai et al., 2016RRID: IMSR_RBRC02332
B6. FVB (Cg)-Tg (Drd1a-cre) EY262Gsat/MmucdGENSAT (Gong et al., 2003)RRID: MMRRC_030989-UCD
B6. FVB (Cg)-Tg (Adora2a-cre) KG139Gsat/MmucdGENSAT (Gong et al., 2003)RRID: MMRRC_036158-UCD
Recombinant DNA
pCAGGS-Myc-MAP2K1 (MEK1)-WTFunahashi et al., 2013N/A
pCAGGS-Myc-MAP2K1 (MEK1)-CAFunahashi et al., 2013N/A
pCAGGS-Myc-MAP2K1 (MEK1)-DNFunahashi et al., 2013N/A
Human CBP cDNAKazusa DNA Research InstituteCat# ORK11778
Mouse Npas4 cDNAYun et al., 2013N/A
pENTR/D-TOPOThermo Fisher ScientificCat# K240020
pDEST15Thermo Fisher ScientificCat# 11802014
pDEST15-CBP-RIDThis paperN/A
pDEST15-CBP-N-TADThis paperN/A
pDEST15-CBP-bramoThis paperN/A
pDEST15-CBP-HATThis paperN/A
pDEST15-CBP-C-TADThis paperN/A
pDEST15-Npas4 1-273 aaThis paperN/A
pDEST15-Npas4 274-597 aaThis paperN/A
pDEST15-Npas4 598-802 aaThis paperN/A
pCAGGS-Myc-CBP-N-TADThis paperN/A
pEF-BOS-GSTThis paperN/A
pEF-BOS-GST-CBP-N-TADThis paperN/A
pEF-BOS-GST-Npas4-390-489 aaThis paperN/A
pEF-BOS-GST-Npas4-490-597 aaThis paperN/A
pEF-BOS-GST-Npas4-598-701 aaThis paperN/A
pcDNA-mGFPThis paperN/A
pcDNA-mGFP-Npas4-WTThis paperN/A
pcDNA-mGFP-Npas4-T423AThis paperN/A
pcDNA-mGFP-Npas4-T427AThis paperN/A
pcDNA-mGFP-Npas4-S577AThis paperN/A
pcDNA-mGFP-Npas4-S580AThis paperN/A
pcDNA-mGFP-Npas4-T611AThis paperN/A
pcDNA-mGFP-Npas4-S615AThis paperN/A
pcDNA-mGFP-Npas4-T423A/T427A/S577A/S580A/T611A/S615A (6A)This paperN/A
pcDNA-mGFP-Npas4-T423E/T427E/S577E/S580E/T611E/S615E (6E)This paperN/A
pcDNA-mGFP-Npas4-274-389 aaThis paperN/A
pcDNA-mGFP-Npas4-390-489 aaThis paperN/A
pcDNA-mGFP-Npas4-490-597 aaThis paperN/A
pcDNA-mGFP-Npas4-598-701 aaThis paperN/A
pcDNA-mGFP-Npas4-701-802 aaThis paperN/A
pcDNA-mGFP-Npas4-390-489 aa-T423AThis paperN/A
pcDNA-mGFP-Npas4-390-489 aa-T427AThis paperN/A
pcDNA-mGFP-Npas4-390-489 aa-T423A/T427AThis paperN/A
pcDNA-mGFP-Npas4-490-597 aa-S577AThis paperN/A
pcDNA-mGFP-Npas4-490-597 aa-S580AThis paperN/A
pcDNA-mGFP-Npas4-490-597 aa-S577A/S580AThis paperN/A
pcDNA-mGFP-Npas4-598-701 aa-T611AThis paperN/A
pcDNA-mGFP-Npas4-598-701 aa-S615AThis paperN/A
pcDNA-mGFP-Npas4-598-701 aa-T611A/S615AThis paperN/A
pGL4.31 [luc2P/GAL4 UAS/Hygro] VectorPromegaCat# C9351
pGL4.15 [luc2P/Hygro] VectorPromegaCat# E6701
pGL4.74 [hRluc/TK] VectorPromegaCat# E6921
pFN11 (BIND) Flexi VectorPromegaCat# C9341
pGL4.15-exon1 BDNF promoterThis paperN/A
pFN11A (BIND)-Npas4-274-802 aaThis paperN/A
pAAV-Sp-CreHikida et al., 2010Nagai et al., 2016N/A
pAAV-Sp-EGFPHikida et al., 2010Nagai et al., 2016N/A
pAAV-CAGGS-Flex-EGFP-P2A-MCS-WPRENagai et al., 2016N/A
pAAV-CAGGS-Flex-EGFP-P2A-MAP2K1-DN-WPRENagai et al., 2016N/A
pAAV- CaMKII-Flex-mCherry-P2A-MCS-WPREThis paperN/A
pAAV- CaMKII-Flex-mCherry-P2A-Npas4-WT-WPREThis paperN/A
pAAV- CaMKII-Flex-mCherry-P2A-Npas4-6A-WPREThis paperN/A
pAAV- CaMKII-Flex-mCherry-P2A-Npas4-6E-WPREThis paperN/A
pAAV- CaMKII-Flex-mCherry-P2A-Npas4-DN-WPREThis paperN/A
pHelperCell BioLabsCat# 340202
pAAV-DJCell BioLabsCat# VPK-420-DJ
Software and Algorithms
Prism 6GraphPadRRID: SCR_002798
Proteome Discoverer 1.4Thermo Fisher ScientificRRID: SCR_014477
MascotMatrix ScienceRRID: SCR_014322
DAVID bioinformatics resourcesHuang da et al., 2009aHuang da et al., 2009bhttps://david.ncifcrf.gov
ZEN Digital Imaging for Light MicroscopyCarl ZeissRRID: SCR_013672
LI-COR Image Studio SoftwareLI-COR BiosciencesRRID: SCR_015795
Gen5 softwareBioTekRRID: SCR_017317
MED-PC IV softwareMED AssociatesRRID: SCR_012156
Adobe Photoshop CS6AdobeRRID: SCR_014199
Adobe Illustrator CS6AdobeRRID: SCR_010279
Other
Identification of CBP-interacting proteins by LC-MS/MSThis paperhttps://dx.doi.org/10.17632/gh228m9gms.2
Identification of phosphorylation site of Npas4 by LC-MS/MSThis paperhttps://dx.doi.org/10.17632/3f5p2p7hbs.2

 Lead Contact and Materials Availability

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Kozo Kaibuchi (kaibuchi@med.nagoya-u.ac.jp).All unique/stable reagents generated in this study will be made available on request but we may require a payment and/or a completed Materials Transfer Agreement if there is potential for commercial application.

 Experimental Model and Subject Details

 Animals

C57BL/6 (RRID: IMSR_JAX:000664) and ICR (RRID: IMSR_JAX:009122) mice were purchased from Japan SLC (Shizuoka, Japan). Npas4 KO (RRID: MGI:3828101) and Npas4 flox (RRID: MGI:3828102) mice on a C57BL/6 genetic background were kindly provided by Dr. Michael E. Greenberg (Harvard Medical School, Boston, MA, USA) and have been previously described (Lin et al., 2008). Drd1-mVenus (C57BL/6J-Tg (mDrd1-YFP) 680-1Koba/KobaRbrc, RRID: IMSR_RBRC03111) and Drd2-mVenus transgenic mice (C57BL/6J-Tg (Drd2-YFP) 364-5Koba/KobaRbrc, RRID: IMSR_RBRC02332) were generated as previously described (Nagai et al., 2016). Drd1a-Cre mice on a C57BL/6 background (B6. FVB (Cg)-Tg (Drd1a-cre) EY262Gsat/Mmucd, RRID: MMRRC_030989-UCD) and Adora2a-Cre mice on a C57BL/6 background (B6. FVB (Cg)-Tg (Adora2a-cre) KG139Gsat/Mmucd, RRID: MMRRC_036158-UCD) were provided by the Mutant Mouse Research and Resource Center (MMRRC) at UC Davis (Davis, CA, USA). Heterozygous Drd1a-Cre and Adora2a-Cre male mice were obtained by crossing heterozygous or homozygous Drd1a-Cre and Adora2a-Cre male mice with C57BL/6 female mice. Mice were housed under a standard 12-h light/dark cycle (light phase 9:00-21:00) at a constant temperature of 23 ± 1°C with free access to food and water throughout the experiments. The male mice used in this study were 7–12 weeks old and 22–29 g body weight unless otherwise indicated. All animal experiments were approved and performed in accordance with the guidelines for the care and use of laboratory animals established by the Animal Experiments Committee of Nagoya University Graduate School of Medicine.

 Cell Lines

COS7 (Species: Cercopithecus aethiops, Sex: male, RRID: CVCL_0224, ATCC, Manassas, VA, USA) and AAV293 (Species: Homo sapiens, Sex: female, RRID: CVCL_KA64, Cell BioLabs, San Diego, CA, USA) cell lines were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Merck, Kenilworth, NJ, USA) containing 10% fetal bovine serum (FBS; Merck). Cells were grown in a humidified atmosphere of 5% CO2 at 37°C.

 Primary Cell Cultures

Primary cultured striatal neurons were prepared from E15-16 mouse embryos of ICR female mice (Japan SLC) as described previously (Cowan et al., 2005) with modifications. The striatal tissues were digested with neuron dissociation solutions (FUJIFILM Wako, Tokyo, Japan) according to the manufacturer’s instructions. Dissociated neurons were seeded on coverslips or dishes coated with poly-D-lysine (PDL; Merck) and cultured in Neurobasal medium (Thermo Fisher Scientific, Waltham, MA, USA) containing 10% FBS (Merck). One hour after plating, the medium was changed to Neurobasal medium containing B-27 supplement (Thermo Fisher Scientific) and 1 mM GlutaMAX (Thermo Fisher Scientific). Neurons were grown in a humidified atmosphere of 5% CO2 at 37°C.

 Method Details

 Materials

The cDNA encoding human CBP (ID: ORK11778) was purchased from Kazusa DNA Research Institute (Chiba, Japan). The cDNA encoding mouse Npas4 (GenBank: BC129861) was obtained as previously described (Yun et al., 2013). Rabbit polyclonal antibody against Npas4 phosphorylated at T427 (anti-pT427 antibody) was produced against the phosphopeptide CTPPYpT427PHQPG (Merck). Rabbit polyclonal antibody against Npas4 phosphorylated at S577, S580 and S615 (anti-pT427, anti-pS577 and pS580 antibodies) were produced against the phosphopeptides CEKLPPpS577PSSPG, CPPSPSpS580PGNGD and CLTPEApS615PVKQS (FUJIFILM Wako, Tokyo, Japan). Rabbit polyclonal anti-Npas4 antibody was produced against GST-Npas4-C-term (Yun et al., 2010). Rabbit polyclonal anti-phospho-CRMP2 (T514) antibody was obtained as previously described (Yoshimura et al., 2005). Rabbit polyclonal anti-phospho-Rasgrp2 (S116/S117) antibody was obtained as previously described (Nagai et al., 2016). The following antibodies and materials were used: rabbit polyclonal anti-CBP (A-22) (RRID: AB_631006 ) and rabbit polyclonal anti-c-Myc (A-14) (RRID: AB_631274 ) antibodies (Santa Cruz, Dallas, TX, USA); Rabbit monoclonal anti-phospho-CREB (S133) (RRID: AB_2561044 ), rabbit monoclonal anti-CREB (RRID: AB_331277 ), mouse monoclonal anti-CREB (RRID: AB_490881 ), rabbit monoclonal anti-Histone H3 (RRID: AB_10544537 ), rabbit monoclonal anti-FosB (RRID: AB_2106903 ), rabbit monoclonal anti-phospho-p44/42 MAPK (ERK1/2) (T202/Y204) (RRID: AB_2315112 ), rabbit monoclonal anti-p44/42 MAPK (ERK1/2) (RRID: AB_390779 ), mouse monoclonal anti-p44/42 MAPK (ERK1/2) (RRID: AB_10695739 ), rabbit monoclonal anti-phospho-SAPK/JNK (T183/Y185) (RRID: AB_823588 ), rabbit polyclonal anti-SAPK/JNK (RRID: AB_2250373 ) rabbit monoclonal anti-phospho-p38 MAPK (T180/Y182), mouse monoclonal p38 MAPK (RRID: AB_331298 ), rabbit monoclonal anti-phospho-AKT (T308) (RRID: AB_2255933 ), rabbit monoclonal anti-AKT (RRID: AB_915783 ) antibodies (Cell Signaling Technology Inc, Danvers, MA, USA); rabbit polyclonal anti-Npas4 (RRID: AB_10674282 ), mouse monoclonal anti-CBP/p300 (RRID: AB_260994 ) antibodies (Merck); rabbit polyclonal anti-GFP (RRID: AB_591819 ) and rabbit polyclonal anti-RFP (RRID: AB_591279 ) antibodies (MBL International, Aichi, Japan); mouse monoclonal anti-GFP antibody (RRID: AB_390913 , Roche, Mannheim, Germany); mouse monoclonal anti-GST antibody (FUJIFILM Wako); rabbit polyclonal anti-Rasgrp2 antibody (RRID: AB_254634 , Thermo Fisher Scientific); mouse monoclonal anti-Jun antibody (RRID: AB_397717 , BD Biosciences, CA, USA); rabbit polyclonal anti-Mkl2 antibody (RRID: AB_10009441 , Novus Biologicals, Littleton, CO, USA); rabbit polyclonal anti-Matrin-3 antibody (RRID: AB_495514 , Bethyl Laboratories, Montgomery, TX, USA); rabbit polyclonal anti-Pur-alpha antibody (RRID: AB_2253242 , Abcam, Cambridge, UK); Alexa 488-, Alexa 555- and Alexa 647- conjugated secondary antibodies against rat, rabbit or mouse immunoglobulin, respectively (RRID: AB_2535794 , RRID: AB_162543 , RRID: AB_2535758 , Thermo Fisher Scientific); active MAPK1 (Carna Biosciences, Kobe, Japan); Okadaic acid (OA) (LC Laboratories, Woburn, MA, USA); Forskolin [7β-Acetoxy-8,13-epoxy-1α,6β,9α-trihydroxy-labd-14-en-11-one], MAP2K1/2 inhibitor U0126 [1,4-diamino-2,3-dicyano-1,4-bis (o-aminophenylmercapto) butadiene], JNK Inhibitor II SP600125 [Anthra [1,9-cd] pyrazol-6 (2H)-one,1,9-pyrazoloanthrone], and PKA inhibitor H-89 Dihydrochloride N-[2-((p-Bromocinnamyl)amino)ethyl]-5-isoquinolinesulfonamide, 2HCl, and PI3K inhibitor LY294002 [2-(4-Morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one] (Merck). p38 MAPK inhibitor SB203580 [4-(4-Fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole] and GSK3 inhibitor SB216763 [3-(2,4-Dichlorophenyl)-4-(1-methyl-1H-indol-3-yl)-1H-pyrrole-2,5-dione] (Tocris Bioscience, Cambridge, UK). Other materials and chemicals were obtained from commercial sources.

 Plasmid Constructs

We subcloned the PCR-amplified cDNA fragments of Npas4 and CBP into pENTR-D/TOPO (Thermo Fisher Scientific). After sequencing, these fragments were further subcloned into appropriate commercial or homemade destination vectors via Gateway technology (Thermo Fisher Scientific) according to the manufacturer’s instructions. The mutants Npas4-T423A, Npas4-T427A, Npas4-S577A, Npas4-S580A, Npas4-T611A, Npas4-S615A, Npas4-6A and Npas4-6E were generated with a PrimeSTAR Mutagenesis Basal Kit (Takara, Shiga, Japan) by changing T423, T427, S577, S580, T611 and/or S615 into alanine or glutamic acid. The exon 1 BDNF promoter region was amplified with primer pair [5′-CCATTAGAGCAAACGCAGTCATA-3′ (forward), 5′ CCACCACTTGGTGTGACTTATG-3′ (reverse)] using mouse genomic DNA and subcloned into the pGL4.15 [luc2P/Hygro] vector (Promega, Madison, WI, USA). pCAGGS-Myc-MAP2K1-WT, -CA and -DN were obtained as previously described (Funahashi et al., 2013). GST-tagged proteins were produced in BL21 (DE3) or Rosetta (DE3) Escherichia coli cells and purified on glutathione-Sepharose 4B beads (GE Healthcare, Chicago, IL, USA). To generate an AAV vector that bicistronically expresses the mCherry protein and the target protein in a Cre-dependent manner, we constructed the pAAV-CaMKII-Flex-mCherry-P2A-MCS-WPRE plasmid by modifying the pAAV-CAGGS-Flex-MCS-WPRE plasmid. We cloned the mCherry cDNA sequence, followed by a self-cleaving 2A peptide sequence and a multiple cloning site (MCS), into the MCS of pAAV-CAGGS-Flex-MCS-WPRE in an inverted manner. The CAGGS promoter was replaced with the CaMKII promoter. Npas4-WT, 6A, 6E or DN (deltaC 274-802 aa) were each cloned into the MCS of the pAAV-CaMKII-Flex-mCherry-P2A-MCS-WPRE plasmid. pAAV-Sp-EGFP, pAAV-Sp-Cre, pAAV-CAGGS-Flex-EGFP-P2A-MCS-WPRE, and pAAV-CAGGS-Flex-EGFP-P2A-MAP2K1-DN-WPRE were generated as previously described (Nagai et al., 2016).

 Laemmli’s SDS-PAGE, Phos-tag SDS-PAGE, and Immunoblotting

Laemmli’s SDS-PAGE was carried out using 6%, 8% or 10% polyacrylamide gels (Nacalai Tesque, Kyoto, Japan). Phos-tag SDS-PAGE was performed with 6%, 7.5% or 10% polyacrylamide gels containing 25 μM Phos-tag acrylamide (FUJIFILM Wako) and 75 μM MnCl2. The proteins were separated via SDS-PAGE and transferred to polyvinylidene difluoride membranes (Immobilon-FL, Merck). The membranes were blocked for 30 min with Blocking-One or Blocking-One P (Nacalai Tesque) and incubated for 1 hr at room temperature or overnight at 4 °C with primary antibodies. After the membranes were washed, they were incubated for 30 min with goat anti-rabbit IgG Alexa Fluor 680 (RRID: AB_2535758 , Thermo Fisher Scientific), goat anti-Mouse IgG, Alexa Fluor 680 (RRID: AB_2535724 , Thermo Fisher Scientific) or anti-mouse IgG DyLight 800 Conjugate (RRID: AB_10693543 , Cell Signaling Technology) at room temperature for 30 min. Specific binding was detected using an infrared (LI-COR Biosciences, Lincoln, NE) imaging system. Band intensities were quantified using ImageStudio software (RRID: SCR_015795 , LI-COR Biosciences).

 In Vitro Phosphorylation Assay

The phosphorylation assay was performed as previously described (Amano et al., 2015Funahashi et al., 2013). Npas4 fragments were expressed in E. coli as GST fusion proteins and purified with glutathione-Sepharose 4B beads (GE Healthcare). The kinase reactions for MAPK1 were performed in 100 μl of a reaction mixture (50 mM Tris/HCl, pH 7.5, 1 mM EDTA, 1 mM EGTA, 1 mM DTT, 5 mM MgCl2, 50 μM [Γ32P] ATP [1-20 GBq/mmol]), 0.05 μM active MAPK1, and 0.3 μM purified GST-Npas4 fragments for 30 min at 30°C. Then, the reaction mixtures were boiled in SDS sample buffer and subjected to SDS-PAGE and silver staining. The radiolabeled proteins were visualized with an image analyzer (FLA9000; GE Healthcare).

 GST Pulldown Assay

For determining the binding site between CBP and Npas4, COS7 cells were transfected GFP-Npas4-WT or Myc-CBP-N-TAD. After 18-24 hr transfection, cells were lysed in lysis buffer [20 mM Tris/HCl, 1 mM EDTA, 150 mM NaCl, 1% NP-40, protease inhibitor cocktail (Roche), and phosphatase inhibitor cocktail (PhosStop, Roche), pH 7.5] and sonicated 3 times for 5 s. After centrifugation at 16,000 × g at 4°C for 10 min, the soluble supernatant was incubated with glutathione-Sepharose 4B beads coated with 100 pmol of GST, GST-CBP or GST-Npas4 fragments for 1 hr at 4°C with gentle rocking. For the interaction assay of exogenous CBP and Npas4, COS7 cells were transfected with the indicated plasmids. After 18-24 hr transfection, cells were pretreated with the indicated kinase inhibitors and then treated with DMSO or OA. Cells were lysed in lysis buffer containing protease inhibitor cocktail and PhosStop, and then sonicated 3 times for 5 s. After centrifugation at 16,000 × g at 4°C for 10 min, the soluble supernatant was incubated in 30 μl of glutathione-Sepharose 4B beads for 1 hr at 4°C with gentle rocking. The beads were washed three times with lysis buffer, eluted by boiling in SDS sample buffer for SDS-PAGE and then subjected to immunoblot analysis with the indicated antibodies.

 Mass Spectrometry

For identification of CBP-interacting proteins, after CPP conditioning for 30 min, brains from 10 mice were rapidly removed and coronal slices (1 mm) were produced using a mouse brain slicer matrix (Brain Science Idea Co., Ltd., Osaka, Japan). The striatum and NAc (approximately +1.54 mm from Bregma) were collected using 3 mm diameter biopsy punch (Miltex, NY, USA) based on the Paxinos and Franklin “the mouse brain in stereotaxic coordinates.” Collected tissues were homogenized with Hypotonic buffer (Nuclear Extract Kit, Active Motif, CA, USA) containing protease inhibitor cocktail and PhosStop using potter homogenizer and incubated on ice for 15 min. The lysate was centrifuged at 800 × g for 10 min at 4°C. The pellet was resuspended with Hypotonic buffer and incubated on ice for 15 min. After addition of a detergent and vortexing for 10 s, the lysate was centrifuged at 16,000 × g for 2 min. The nuclear pellet was extracted in lysis buffer containing protease inhibitor cocktail and PhosStop, and then sonicated 3 times for 10 s. The lysate was centrifuged at 16,000 × g for 10 min at 4°C. The supernatant was used as the nuclear extracts. A total of 250 pmol of GST or GST-CBP-N-TAD, immobilized on glutathione Sepharose 4B beads, was incubated with the nuclear extracts for 1 hr at 4°C with rotation. The beads were then washed three times with Lysis buffer and an additional three times with wash buffer (20 mM Tris/HCl, 1 mM EDTA, and 150 mM NaCl, pH 7.5) to remove the detergent from the beads.For identification of Npas4 phosphorylation site, GST-Npas4 fragment was expressed in COS7 cells with or without the coexpression of MAP2K1-CA. Cells were lysed in lysis buffer containing protease inhibitor cocktail (Roche) and PhosStop (Roche), and sonicated 3 times for 5 s. After centrifugation at 16,000 × g at 4°C for 10 min, the soluble supernatant was incubated in 30 μl of glutathione-Sepharose 4B beads for 1 hr at 4°C with rotation. The beads were then washed three times with lysis buffer and an additional three times with wash buffer.The bound proteins were extracted from the beads using urea solution (1 M urea and 50 mM NH4HCO3), reduced via incubation in 5 mM dithiothreitol for 30 min, and alkylated using 10 mM iodoacetamide for 1 hr in the dark. The proteins were digested with Trypsin/Lys-C (Promega) or Glu-C (Promega)/Asp-N (FUJIFILM Wako). Demineralization was performed using SPE c-tips (Nikkyo Technos, Tokyo, Japan) according to the manufacturer’s instructions. The peptides were analyzed by LC−MS using an Orbitrap Fusion mass spectrometer (Thermo Fisher Scientific Inc) coupled to an UltiMate3000 RSLCnano LC system (Dionex Co., Amsterdam, the Netherlands) using a nano HPLC capillary column, 150 mm × 75 m i.d (Nikkyo Technos Co., Tokyo, Japan) via a nanoelectrospray ion source.Reversed-phase chromatography was performed with a linear gradient (0 min, 5% B; 100 min, 40% B) of solvent A (2% acetonitrile with 0.1% formic acid) and solvent B (95% acetonitrile with 0.1% formic acid) at an estimated flow rate of 300 nl/min. A precursor ion scan was carried out using a 400-1600 mass to charge ratio (m/z) prior to MS/MS analysis. Tandem MS was performed by isolation at 0.8 Th with the quadrupole, HCD fragmentation with normalized collision energy of 30%, and rapid scan MS analysis in the ion trap. Only those precursors with charge states of 2-6 were sampled for MS2. The dynamic exclusion duration was set to 10 s with a 10 ppm tolerance. The instrument was run in top speed mode with 3 s cycles.

 Data Analysis for Mass Spectrometry

The raw data were processed using Proteome Discoverer 1.4 (RRID: SCR_014477 , Thermo Fisher Scientific) in conjunction with the MASCOT search engine, version 2.6.0 (RRID: SCR_014322 , Matrix Science Inc., Boston, MA) for protein identification. Peptides and proteins were identified against the mouse protein database in UniProt (release 2018_11), with a precursor mass tolerance of 10 ppm and a fragment ion mass tolerance of 0.8 Da. Peptides were identified with the false discovery rate (FDR) < 1% (Identification of phosphorylation site of Npas4) or 5% (Screening of CBP-interacting protein). Fixed modification was set to cysteine carbamidomethylation, and variable modifications were set to methionine oxidation and serine, threonine tyrosine phosphorylation. Two missed cleavages by trypsin/Lys-C or Glu-C/Asp-N were allowed. Quantification was carried out by SIM analysis, to obtain the peak area values of dual phosphorylated peptides. Gene ontology (GO) analysis (biological process) and KEGG pathway analysis were performed using the DAVID bioinformatics resources (https://david.ncifcrf.gov)

 Immunofluorescence Analysis

Mice were anesthetized with 2,2,2-tribromoethanol/tert-amyl alcohol solution (Avertin, 200 mg/kg, i.p.) (FUJIFILM Wako) for rapid and deep anesthesia and transcardially perfused with 4% paraformaldehyde. Then, the brains were removed and incubated in 4% paraformaldehyde overnight at 4 °C. The brains were sectioned coronally with a vibratome (VT1200S, Leica Microsystems, Wetzlar, Germany) at a thickness of 50 μm. The brain slices were incubated with 0.1% Triton X-100/phosphate-buffered saline (PBS) for 10 min and blocked with Blocking-One P for 30 min. Cultured striatal neurons were transfected with GFP-Npas4-WT at 7 days in vitro (DIV) using the Lipofectamine 2000 reagent (Thermo Fisher Scientific) according to the manufacturer’s instructions. Neurons were treated with DMSO or 10 μM forskolin for 30 min at 8 DIV and fixed with 3.7% formaldehyde in PBS for 10 min at room temperature. Neurons were treatment with 0.05% Triton X-100/PBS for 10 min on ice and blocked with 10% donkey serum albumin in PBS. The brain slices and neurons were incubated with each indicated antibody overnight at 4°C or for 1hr at room temperature. After washing, the samples were incubated with Alexa Fluor 488-, Alexa Fluor 555- or Alexa Fluor 647-conjugated secondary antibodies for 1 hr. Nuclei were visualized by staining with Hoechst 33342 (Nacalai). Confocal images were recorded with LSM 780 microscopes built around an Axio Observer Z1 with Plan-Apochromat 20 × (numerical aperture [NA] 0.8), C-Apochromat 40 × (NA 1.2) or Plan-Apochromat 63 × (NA 1.40) lenses under the control of ZEN Digital Imaging for Light Microscopy (RRID: SCR_013672 , Carl Zeiss, Oberkochen. Germany). The entire image of a coronal brain section was observed with a fluorescence microscope (BZ-9000 and BZ-X800, Keyence, Osaka, Japan).

 Dual-Luciferase Reporter Assay

Striatal neurons were seeded on 24-well plates and cultured for 7-8 days. Neurons were cotransfected with pGL4-exon1 BDNF promoter or pGL4.31[luc2P/GAL4 UAS/Hygro] vector (Promega) and expression vectors using the Lipofectamine 2000 transfection reagent (Thermo Fisher Scientific) according to the manufacturer’s instructions. As necessary, an empty control plasmid was added to ensure that each transfection received the same amount of total DNA. To normalize for transfection efficiency, we added pRluc-TK (Renilla luciferase) reporter plasmid (Promega) to each transfection. Luciferase activities were measured using a dual luciferase reporter assay system (Promega) and a PowerScan4 microplate reader (Bio Tek, Winooski, VT, USA). The data were analyzed using Gen5 software (RRID: SCR_017317 , Bio Tek).

 AAV Preparation and Injection

AAV vectors were prepared and titered as described previously (Sooksawate et al., 2013). Briefly, plasmids for the AAV vector, pHelper (Cell BioLabs), and pAAV-DJ (Cell BioLabs) were transfected into AAV293 cells. After 3 days of incubation, cells were collected and purified. The titers of AAV were estimated by qPCR. Mice were anesthetized with Avertin and positioned in a stereotaxic frame (David Kopf, Tujunga, CA, USA). An AAV virus (0.5 μL, 1.0 × 1012 genome copies/mL) was injected into the NAc through a glass microinjection capillary tube at a 10° angle and a rate of 0.1 μL/min (0.5 μL/site, four sites). The anteroposterior, mediolateral, and dorsoventral coordinates relative to bregma were as follows (in mm): +1.6, ± 1.5, and −4.4; and +1.0, ± 1.6, and −4.5.

 Behavioral Analysis

The conditioned place preference test was performed as previously described (Nagai et al., 2016). The apparatus consisted of two compartments: a transparent Plexiglas box and a black Plexiglas box (W 16 cm × D 16 cm × H 17 cm). To be used as cues to enable the mice to distinguish between the boxes, the floor of the transparent box was covered with uneven Plexiglas and that of the black box was covered with smooth black Plexiglas. All sessions were conducted under conditions of dim illumination (40 lux). The place-conditioning schedule consisted of three phases: preconditioning, conditioning, and postconditioning. In the preconditioning phase, the mouse was allowed to move freely between the boxes for 30 min, twice a day. During the second session on day 1, the time spent in each of the boxes was measured for 15 min using MED-PC IV software (RRID: SCR_012156 , Brainscience Idea, Osaka, Japan). Conditioning was counterbalanced between compartments to ensure that the procedure was unbiased. No initial place preference or aversion to either compartment was observed in the experiment. For 3 days, the mice were conditioned to an intraperitoneal saline injection in one chamber for 30 min during a morning session and to a 10 mg/kg intraperitoneal cocaine injection in the opposite chamber for 30 min during an afternoon session. Control animals received saline during every session. On day 5, the postconditioning phase was performed in a manner that was similar to the preconditioning phase, i.e., the mice were allowed free access to both compartments for 15 min, and the time spent in each of the boxes was measured. CPP was calculated for each mouse as the difference in the time spent on the cocaine-conditioned side between the pre- and postconditioning phases.

 Quantification and Statistical Analysis

Data analysis was performed using Prism 6 Statistics software (RRID: SCR_002798 , GraphPad Software, Inc., La Jolla, USA). All data are expressed as the means ± standard error of the mean (SEM). One-way or two-way analysis of variance (ANOVA) was used, followed by Tukey’s or Dunnett’s multiple-comparison test when the F ratios were significant (p < 0.05). Differences between two groups were tested for significance using t test.

Dr. Raymond Oenbrink