bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Cocaine-dependent acquisition of locomotor sensitization and conditioned place preference requires

2 D1 dopaminergic signaling through a cyclic AMP, NCS-Rapgef2, ERK and Egr-1/Zif268 pathway

3 Abbreviated title: NCS-Rapgef2 in cocaine rewarding

4

5 Sunny Zhihong Jiang1, 4, Sean Sweat1, 4, Sam Dahlke1, Kathleen Loane1, 4, Gunner Drossel1, Wenqin Xu1,

6 Hugo A. Tejeda2.4, Charles R. Gerfen3, and Lee E. Eiden1,4*

7 1Section on Molecular Neuroscience, 2Unit on Neuromodulation and Synaptic Integration, 3Laboratory of Systems

8 Neuroscience and 4Dendritic Dynamics Hub, National Institute of Mental Health Intramural Research Program,

9 Bethesda, MD, USA 20892

10

11 *To whom correspondence should be addressed: Lee E. Eiden, Section on Molecular Neuroscience, Na tional Institute

12 of Mental Health Intramural Research Program, 9000 Rockville Pike, Building 49, Room 5A38, Bethesda, MD, USA,

13 20892, Tel.: (301) 496-4110, E-mail, [email protected]

14 15 Number of pages: 46 pages of main text 16 Number of figures: 9 17 Number of words for Abstract: 244 18 Number of words for Significance Statement: 120 19 Number of words for Introduction: 649 20 Number of words for Discussion: 1500 21 Extended data: 5 figures 22 23 CONFLICT OF INTEREST

24 The authors declare no competing financial interests.

25 26 ACKNOWLEDGEMENTS

27 Thanks to Haiying Zhang for mouse colony management and genotyping. Thanks to Dr. Ka zu Na kazawa (The

28 Southern Research Institute) for helpful comments. This work was supported by the National Institute of Mental

29 Health Intramural Research Program, Project 1ZIAMH002386 to L.E.E.

30

1

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 ABSTRACT

2 Elucidation of the underlying mechanism of dopamine signaling to ERK that underlies plasticity

3 in dopamine D1 receptor expressing neurons leading to acquired cocaine preference is incomplete.

4 NCS-Rapgef2 is a novel cAMP effector, expressed in neuronal and endocrine cells in adult

5 mammals, that is required for D1 dopamine receptor-dependent ERK phosphorylation in mouse

6 brain. In this report, we studied the effects of abrogating NCS-Rapgef2 expression on cAMP-

7 dependent ERKEgr-1/zif268 signaling in cultured neuroendocrine cells; in D1 medium spiny

8 neurons (MSNs) of nucleus accumbens slices; and in mouse brain in a region-specific manner.

9 NCS-Rapgef2 deletion in the nucleus accumbens (NAc) in adult mice, using AAV-mediated

10 expression of cre recombinase, eliminated cocaine-induced ERK phosphorylation and Egr-

11 1/Zif268 upregulation in D1-MSNs and cocaine-induced behaviors including locomotor

12 sensitization and conditioned place preference (CPP). Abrogation of NCS-Rapgef2 gene

13 expression in medium prefrontal cortex and basolateral amygdala, by crossing mice bearing a

14 floxed Rapgef2 allele with a cre mouse line driven by calcium/calmodulin-dependent kinase IIα

15 promoter also eliminated cocaine-induced phospho-ERK activation and Egr-1/Zif268 induction,

16 but without effect on the cocaine-induced behaviors. Our results indicate that NCS-Rapgef2

17 signaling to ERK in dopamine D1-receptor expressing neurons in the NAc, but not in corticolimbic

18 areas, contributes to cocaine-induced locomotor sensitization and CPP. Ablation of cocaine-

19 dependent ERK activation by elimination of NCS-Rapgef2 occurred with no effect on

20 phosphorylation of CREB in D1 dopaminoceptive neurons of NAc. This study reveals a new

21 cAMP-dependent signaling pathway for cocaine-induced behavioral adaptations, mediated

22 through NCS-Rapgef2/phospho-ERK activation, independently of PKA/CREB signaling.

23 24

2

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 SIGNIFICANCE STATEMENT 2 ERK phosphorylation in dopamine D1 receptor expressing neurons exerts a pivotal role in

3 psychostimulant-induced neuronal gene regulation and behavioral adaptation, including locomotor

4 sensitization and drug preference in rodents. In this study, we examined the role of dopamine

5 signaling through the D1 receptor via a novel pathway initiated through the cAMP-activated

6 guanine nucleotide exchange factor NCS-Rapgef2 in mice. NCS-Rapgef2 in the nucleus

7 accumbens is required for activation of ERK and Egr-1/Zif268 in D1 dopaminoceptive neurons

8 after acute cocaine administration, and subsequent enhanced locomotor response and drug seeking

9 behavior after repeated cocaine administration. This novel component in dopamine signaling

10 provides a potential new target for intervention in psychostimulant-shaped behaviors, and new

11 understanding of how D1-MSNs encode the experience of psychomotor stimulant exposure.

12

3

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 INTRODUCTION

2 Behavioral plasticity, including locomotor sensitization and development of drug

3 preference, is associated with psychomotor stimulant-induced neuroadaptation in brain reward

4 circuits linked to addiction (Robinson and Berridge, 2000; Laakso et al., 2002; Fernando and

5 Robbins, 2011; Uhl et al., 2019). Neurons in striatum, prefrontal cortex, amygdala and

6 hippocampus that receive dopaminergic input from the ventral tegmental area (VTA) are central

7 to psychomotor stimulant-induced neuroadaptation (Rebec and Sun, 2005; Nestler and Luscher,

8 2019). Amphetamine and cocaine increase the concentration of dopamine released from nerve

9 terminals in these brain regions (Heien et al., 2005; Nestler and Luscher, 2019). Dopamine acts at

10 receptors promoting either an increase (D1 type) or a decrease (D2 type) in cyclic AMP production

11 in distinct dopaminoceptive neurons. A balance between these two processes contributes to the

12 behavioral sequelae, including addiction, of repeated psychomotor stimulant use (Lobo and

13 Nestler, 2011). Cellular mechanisms for the behavioral effects of psychomotor stimulant exposure

14 have been sought by examining dopamine signaling through the Gs-coupled D1 receptor in various

15 brain areas. One of these is the nucleus accumbens (NAc), which contains GABAergic medium

16 spiny neurons (MSNs) neurons whose activity is modulated by dopamine, and represent the

17 afferent targets of the reward pathway from VTA to NAc in mammalian brain (Hikida et al., 2010;

18 Russo and Nestler, 2013).

19 The MAP kinase ERK is activated in vivo upon treatment with D1 agonists, cocaine or

20 amphetamine (Valjent et al., 2000; Gutierrez-Arenas et al., 2014; Jin et al., 2019) in brain regions

21 innervated by dopaminergic neurons. Pharmacological and genetic manipulations have further

22 implicated ERK activation in transcriptional effects of psychomotor stimulants, notably expression

23 of the immediate-early gene egr1/zif268, linked to psychomotor stimulant-induced behaviors

4

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 (Valjent et al., 2006c). However, genetic and pharmacological tools have limited cellular and

2 anatomical specificity for analysis of these pathways. For example, generalized effects on

3 locomotor activity and co-regulatory effects of ERK1 and ERK2 complicate interpretation of

4 ERK1/2 knockout effects on both behavior and cell signaling (Mazzucchelli et al., 2002; Ferguson

5 et al., 2006; Hutton et al., 2017). Consequently, it has been difficult to identify a signaling pathway

6 arising from D1 receptor activation leading to the specific ERK activation and Egr1/Zif268

7 induction that mediates cellular plasticity within dopaminoceptive neurons, in defined brain

8 circuits, responsible for the altered behaviors typical of repeated psychomotor stimulant

9 administration.

10 In particular, the role of kinase A (PKA) as the downstream effector of cyclic AMP

11 in the D1-dependent activation of Egr-1/Zif268 by ERK (Funahashi et al., 2019), leading to

12 cocaine-dependent locomotor sensitization, has been difficult to confirm. This suggests that a

13 second cAMP-dependent pathway for ERK regulation of Egr-1/Zif268 induction, and subsequent

14 behavioral effects of cocaine mediated through dopamine, may exist in D1-MSNs.

15 We have previously identified a novel pathway for ERK activation via the novel cAMP

16 effector NCS-Rapgef2 in NS-1 neuroendocrine cells in culture (Emery and Eiden, 2012; Emery et

17 al., 2013; Emery et al., 2014; Emery et al., 2017a). NCS-Rapgef2 is activated by cAMP binding

18 to facilitate GDP-GTP exchange on the Rap1 molecule, allowing Rap1 to further stimulate the

19 MAP kinase cascade culminating in ERK phosphorylation/activation (see Jiang et al., 2017). Using

20 this assay as a read-out for the NCS-Rapgef2 dependent cAMP pathway, we have shown that the

21 dopamine D1 receptor couples to NCS-Rapgef2 to activate ERK in neuroendocrine cells, and

22 further demonstrated that this pathway is required for D1-dependent activation of ERK in

23 hippocampus and other forebrain regions of the adult mouse brain in vivo (Jiang et al., 2017).

5

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 These data open the possibility that a direct pathway for activation of ERK by the Gs/olf-

2 coupled D1 receptor leading to Egr-1/Zif268 induction exists in D1 dopaminoceptive neurons

3 involved in the behavioral effects of repeated cocaine administration. Here, we have asked whether

4 the NCS-Rapgef2-mediated activation of ERK after cyclic AMP elevation by the Gs-coupled D1

5 receptor is responsible for the behavioral effects of psychomotor stimulant administration, and if

6 so, in which neuronal ensembles or circuits it might be relevant.

7

6

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 METHODS AND MATERIALS

2 Animals and drugs administered in vivo

3 Mice (wild-type or transgenic) on C57BL6J background were housed 2-5 per cage and

4 acclimatized to 12-hour light/12-hour dark cycle with food and water ad libitum. Animal care,

5 drug treatment and surgeries were approved by the NIMH Institutional Animal Care and Use

6 Committee (ACUC) and conducted in accordance with the NIH guidelines. The floxed Rapgef2

7 mouse strain Rapgef2cko/cko and corticolimbic Rapgef2 KO mouse strain Camk2α-cre+/-

8 ::Rapgef2cko/cko were generated as described previously (Jiang et al., 2017). The transgenic Drd1a-

9 tdTomato BAC mouse (The Jackson Laboratory, #016204), expressing tdTomato selectively in

10 D1-dopaminoceptive neurons (Shuen et al., 2008), was obtained from the Jackson Laboratory.

11 Pharmaceutical grade D1 dopamine receptor agonist SKF81297, D2 antagonist eticlopride, and

12 cocaine were purchased from Sigma (St. Louis, MO) and dissolved in saline. All drugs used in

13 vivo were administrated by intraperitoneal (i.p.) injection.

14

15 Brain slice pharmacology

16 250 µm thick coronal slices of mouse brain containing the nucleus accumbens were

17 prepared as described previously for electrophysiological examination under perfusion (Tejeda et

18 al., 2017), but were instead incubated in a static bath of continuously aerated artificial

19 cerebrospinal fluid, total volume 10 ml per slice, to which various drugs and drug combinations

20 were added to examine the parameters of dopamine and cAMP-dependent signaling in

21 dopaminoceptive cells of core and shell of this brain region. Inhibitors or vehicle were added for

22 a 10 min period prior to addition of 10 μM SKF81297, or vehicle. Incubation at 34°C was

23 continued for 30 min, and slices then quickly removed to cold 10% formalin in phosphate-buffered

7

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 saline for immunohistochemical staining and microscopical analysis as described below. N6-

2 phenyl, 9-tetrahydrofuranyl adenine (EL1101), a preferential inhibitor of the Rap guanine

3 nucleotide exchange activity of NCS-Rapgef2 compared to cAMP effectors protein kinase A and

4 Epac , was synthesized and purified as described previously (Emery et al., 2017b). All drugs were

5 initially dissolved in DMSO, present to a final concentration of 0.1% in all experiments. Final

6 concentration of U0126 was 10 µM, and final concentration of EL1101 was 100 µM, in slice

7 experiments.

8

9 Cell lines and cell culture

10 NS-1 cells (Emery et al., 2013) were cultured in Roswell Park Memorial Institute (RPMI)

11 1640 medium supplemented with 10% HyClone horse serum (GE Healthcare, Marlborough, MA),

12 5% heat-inactivated fetal bovine serum, 2 mM L-glutamine, 100 U/ml penicillin, and 100 µg/ml

13 streptomycin. The established NS-1 cell line with NCS-Rapgef2 knocked-out was described

14 previously (Jiang et al., 2017). The cell-permeable cyclic AMP analog 8-chlorophenylthio-cyclic

15 AMP (8-CPT-cAMP) was purchased from Biolog Life Science Institute (Bremen, Germany) and

16 the stock solution was prepared at 100 mM in DMSO, diluted in culture media to a final

17 concentration of 100 μM.

18

19 Viral injection

20 Modified AAV vectors were obtained from Penn Vector Core. Rapgef2 cko/cko mice were

21 bilaterally injected with AAV9.hSynap.HI.eGFP-Cre.WPRE.SV40, encoding eGFP-fused Cre

22 recombinase under the control of the human synapsin promoter, to knock out Rapgef2 expression

23 in nucleus accumbens on both sides of the brain. AAV9.hSynap.eGFP.WPRE.SV40, encoding

8

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 eGFP without Cre, also under the control of the synapsin promoter, was used as a control. Surgeries

2 and viral injection were conducted according to the “NIH-ARAC guidelines for survival rodent

3 surgery”. Briefly, animals were anesthetized, shaved and mounted into the stereotaxic apparatus.

4 A small midline incision was then made and the pericranial tissue was teased away from the skull

5 with an ethanol-soaked swab to enable identification of the Bregma and Lambda areas. Using a

6 small hand-held drill, a very small hole was made in the skull according to the calculated

7 coordinates for nucleus accumbens (Bregma 1.5 mm, ML 0.7 mm, DV 3.8 mm). A Hamilton

8 syringe (pre-loaded with viral particles) was then slowly lowered, penetrating the dura to the

9 determined depth. A volume of 0.3 μl of the virus (~0.5x109 infectious particle per microliter) was

10 slowly (~0.1 μl/min) injected into the brain. A 2~3-minute wait is performed before the needle is

11 very slowly retracted from the brain to prevent backflow of the viral vector. The animals were

12 allowed to recover and subjected to behavioral tests four weeks after viral injection. Brain injection

13 sites were histologically verified with counterstaining from post-mortem sections and plotted on

14 standardized sections from the stereotaxic atlas.

15

16 Immunohistochemistry

17 Immunohistochemistry was conducted as previously described (Jiang et al., 2017) after animal

18 perfusion with 4% paraformaldehyde. Mouse brains were sectioned by Vibratome at a 40 μm

19 thickness. Free-floating sections were washed in TBS containing 0.5% Triton X-100 (TBST; 3

20 washes, 15 min), incubated at room temperature in blocking solution (10% normal goat or donkey

21 serum in PBST; 1h), and then incubated in primary antibody diluted in blocking solution overnight

22 at 4°C. The following day sections were washed in TBST (3 washes, 15 min), incubated in the

23 dark in Alexa 555-conjugated goat anti-rabbit-IgG, Alexa 488-conjugated goat anti-mouse IgG,

9

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Alexa 488-conjugated goat anti-rabbit-IgG, Alexa 555 -conjugated donkey anti-goat-IgG or Alexa

2 488-conjugated donkey anti-rabbit-IgG (1:300; Life Technologies, Grand Island, NY) for 2 h

3 following primary antibody incubation. Sections were mounted in Vectashield (Vector

4 Laboratories, Burlingame, CA). The primary antibodies used were rabbit anti-pERK (1:1500, Cell

5 Signaling, Danvers, MA), anti-pCREB (1:1000, Cell Signaling, Denvers, MA), mouse anti-PSD95

6 (1:1000, NeuroMab, Davis, CA), mouse anti-Bassoon (1:250, Enzo Life Sciences, Inc,

7 Farmingdale, NY), goat anti-tdTomato (1:1000, MyBiosource, Inc, San Diego, CA), rabbit anti-

8 Egr1 (588) (1:7500, Santa Cruz, CA), rabbit anti-Rapgef2 (NNLE-2, custom-made by Anaspec,

9 see ref. (Jiang et al., 2017). Confocal images were obtained on a Zeiss LSM 510 confocal

10 microscope at the National Institute of Neurological Disorders and Stroke Light Imaging Facility.

11 Immunoreactive (IR) signals of NCS-Rapgef2, phospho-ERK and Egr-1 from different brain areas

12 as indicated were quantified by NIH Image J using the mean gray values of integrated density after

13 being converted to gray scale. To quantify the NCS-Rapgef2, phospho-ERK and phospho-CREB

14 IR signals in AAV viral targeted neurons in the NAc, confocal images with double fluorescence

15 channels were imported into Fiji ImageJ and split into different channels. Images in GFP channel

16 were inverted and made into binary mask after adjusting threshold. GFP positive neurons were

17 selected as ROI following particle analysis. The ROI selections were overlaid to the NCS-Rapgef2,

18 phospho-ERK or phospho-CREB channel. Cell number for phospho-ERK positive neurons or

19 mean gray value for NCS-Rapgef2, phospho-CREB IR in ROIs created from GFP positive neurons

20 were measured. GFP signal from AAV-hSyn-eGFP control viral injection and AAV-hSyn-cre-

21 eGFP have different ROIs since cre-eGFP is localized to nuclei and eGFP distributes throughout

22 the cell. pCREB signal is localized to nuclei, so ROI selections from cre-eGFP and eGFP can both

23 completely cover pCREB signal for GFP+/pCREB+ double positive neurons, while this is not the

10

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 case for GPF+/phospho-ERK (pERK) double positive neurons. Since all neurons show basal

2 pCREB expression, quantification of pCREB is more accurate using intensity. However, pERK

3 signal distributes throughout the cell, so that ROI selections from cre-eGFP do not completely

4 cover pERK signal for the GFP+/pERK+ double positive neurons. Thus, more accurate

5 quantification of pERK/GPF co-expression was more accurately assessed via counting of cell

6 number.

7 Western blot for cultured cell and mouse tissues

8 NS-1 cells were seeded in collagen 1-coated 12-well plates and collected in ice-cold RIPA

9 lysis buffer (Thermo Scientific, Rockford, IL) containing 1x Halt™ Protease and Phosphatase

10 Inhibitor Cocktail (Thermo Scientific). Tissues were punched from 0.5 mm coronal sections of

11 mouse brains, weighed, and snap-frozen. Samples were sonicated on ice with RIPA buffer

12 supplemented with Halt Protease and Phosphatase Inhibitor Cocktail (Thermo Scientific). RIPA-

13 insoluble fractions were removed by centrifugation (3000 rpm for 10 min at 4°C). Supernatants

14 were retained, and protein concentration was determined by MicroBCA Protein Assay kit (Thermo

15 Scientific) according to the protocol provided by the manufacturer. Western blot was performed

16 as described previously (Jiang et al., 2017). The rabbit monoclonal antibodies including anti-ERK

17 (Cell Signaling Technology, Cat# 9102, 1:1000) , rabbit anti-phospho-ERK (Cell Signaling

18 Technology, Cat# 9101, 1:1000), rabbit anti-Egr-1 (15F7)(Cell Signaling Technology, Cat# 4153;

19 1:1000) and anti-GAPDH (D16H11) (Cell Signaling Technology, Cat# 5174; 1:1000), were all

20 purchased from Cell signaling Technology and used in the Western blot according to the

21 manufacturer’s recommendations. Immuoreactive bands were visualized with a SuperSignal West

22 Dura Chemiluminescent Substrate (Thermo Scientific), photographed with a ChemiDoc Imaging

23 system, and quantified with NIH ImageJ.

11

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1

2 Behavioral tests

3 Elevated zero maze The elevated zero maze for anxiety was performed by placing the mouse into

4 an open quadrant of a continuous circular 5.5 cm-wide track elevated 65 cm above the floor and

5 divided into alternating walled and open quadrants in a dimly lit room, for 6 min. Video recording

6 from above in the last 5 min was automatically scored for time spent in each quadrant (Top Scan

7 software suite, Clever System Inc.).

8 Locomotor sensitization The mice were tested for sensitized responses to cocaine using a two-

9 injection protocol (Valjent et al., 2010). Briefly, mice were placed in an Accuscan open field

10 chamber (20 X 20 cm2, 8 beam sensors for each dimension) to monitor locomotor activity. Room

11 light was set up as 30-50 lux. For the first 4 days, mice were placed for 30 min in the activity

12 chamber (habituation phase), received an injection of saline (i.p.), then placed back to the chamber

13 for 1 h. Then locomotor sensitization was tested by replacing injection of saline with cocaine (15

14 mg/kg, i.p.) for two continuous days in the same environment.

15 Conditioned Place Preference Place-conditioning boxes consisted of two chambers with balanced

16 contexts. A partition with an opening separated the two chambers but allowed animals access to

17 either side. This partition was closed off during the pairing days. The walls, floors and partition of

18 chambers were cleaned with CLIDOX-S and dried after each session. The conditional place

19 preference paradigm was performed using a biased strategy as follows: On day 1, mice were

20 allowed to explore both sides for 15 min, and time spent in each side was recorded. Animals were

21 separated into groups with approximately equal biases for the sides to start from. The side on which

22 an animal spent most time was defined as “preferred side” for this mouse. On day 2, starting from

23 the unpreferred side, animals were allowed to explore both sides for 15 mins, and data were

12

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 collected to calculate place preference for the preconditioning phase. Beginning on Day 3, the

2 animals were paired for 8 days (30 min per day), with the saline group receiving injections (0.9%

3 sodium chloride) on both sides of the boxes; whereas the drug-paired group received cocaine (10

4 mg/kg) on the unpreferred side and saline on the preferred side. On day 11, all the animals were

5 given a saline injection, placed on the unpreferred side, and allowed to run freely between the two

6 sides for 15 min, and time spent on each side was recorded (Top Scan software suite, Clever

7 System Inc.).

8

9 Statistics

10 Statistical analyses were conducted using Sigma Plot 14.0. Student’s t-tests and factorial

11 model analysis of variances (ANOVA) were employed where appropriate. Post hoc analyses were

12 performed using Bonferroni Test or Fisher’s LSD. Data were presented as mean ± s.e.m.

13 Differences were considered to be significant when p<0.05.

14

15 RESULTS

16 NCS-Rapgef2 dependency of phospho-ERK activation and Egr-1 induction in response to

17 cAMP elevation in NS-1 cells

18 The immediate early gene Egr-1/Krox-24/Zif268 is induced by cocaine treatment in vivo,

19 and implicated in the behavioral actions of cocaine (Valjent et al., 2006c). We have previously

20 shown that Egr-1 and NCS-Rapgef2 are both required for neuritogenesis induced by Gs-coupled

21 GPCRs including the D1 receptor, or elevation of cAMP itself, in neuroendocrine cells (Ravni et

22 al., 2008; Emery et al., 2013; Emery et al., 2017a; Jiang et al., 2017). In order to link cAMP-

23 dependent activation of NCS-Rapgef2 to signaling to ERK resulting in Egr-1 protein induction,

13

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 we treated NS-1 rat pheochromocytoma cells, and an NS-1 cell line in which Rapgef2 expression

2 was deleted with CRISPR-mediated gene mutation (Jiang et al., 2017), with 100 µM of the cell-

3 permeant cAMP analog 8-CPT-cAMP, and measured levels of phospho-ERK (pERK) and Egr-1.

4 ERK phosphorylation and Egr-1 induction in response to 8-CPT-cAMP were abolished in the

5 absence of NCS-Rapgef2 (Figure 1). This prompted us to examine whether elevation of both pERK

6 and Egr-1 after cocaine administration, in dopaminoceptive neurons in vivo, might also be linked

7 by the cAMP-dependent action of NCS-Rapgef2.

8

9 Postsynaptic localization of NCS-Rapgef2 in dopaminoceptive regions of mPFC, BLA, and

10 NAc

11 NCS-Rapgef2 is highly expressed in corticolimbic areas and striatum in mouse brain,

12 including cell types and brain regions (cortical pyramidal cells; hippocampal granule cells of

13 dentate gyrus and pyramidal cells of CA1-3; basolateral amygdalar excitatory neurons; central

14 amygdalar inhibitory neurons; and NAc medium spiny neurons (MSNs) (Jiang et al., 2017)) which

15 receive innervation from dopaminergic neurons of the VTA and substantia nigra. Modulation of

16 neurons in these brain regions by dopamine is implicated in several aspects of learning, memory,

17 stress responding, and reward, and dysfunction of these neurons is similarly implicated in the

18 pathogenesis of a broad range of neuropsychiatric and neurological disorders including

19 Parkinson’s disease, schizophrenia, and drug addiction. We wished to determine if NCS-Rapgef2

20 was localized within dopaminoceptive neurons at an intracellular location consistent with post-

21 synaptic responses to dopamine receptor activation on cells in these regions. For this purpose, we

22 examined the co- and differential localization of NCS-Rapgef2 and both pre- and post-synaptic

23 markers in these regions. NCS-Rapgef2 immunoreactive (IR) signal is not colocalized with

14

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Bassoon, a scaffolding protein of presynaptic active zone, but rather is in close apposition to the

2 Bassoon puncta in mPFC, NAc and BLA (Figure 2A). However, NCS-Rapgef2 IR signal is

3 colocalized with postsynaptic marker PSD95 IR signal in the soma and dendrites of neurons in

4 mPFC pyramidal cells, NAc medium spiny neurons and excitatory neurons in BLA (Figure 2B).

5 These results suggest a post-synaptic location of NCS-Rapgef2 in neurons including those

6 responsive to dopamine.

7

8 ERK activation by D1 receptor stimulation in mouse nucleus accumbens ex vivo: dependence

9 on NCS-Rapgef2.

10 Given the existence of a cyclic AMPERKEgr-1/Zif268 pathway dependent on NCS-

11 Rapgef2 in neuroendocrine cells in culture, we next inquired about the existence of this pathway

12 in neurons receiving dopaminergic input within the mouse CNS, as already implied by our

13 previous observations of Rapgef2-dependent ERK activation in various brain regions after

14 systemic administration of SKF81297 (Jiang et al., 2017). Accordingly, we examined the effects

15 of SKF81297 on pERK activation after its direct application to nucleus accumbens-containing

16 brain slices, in the presence and absence of the NCS-Rapgef2 inhibitor 6-phenyl-9-

17 tetrahydrofuranyl-adenine (EL1101). 10 μM SKF81297 produced a robust increase in pERK in

18 both core and shell of nucleus accumbens slices in a pattern similar to that seen after SKF81297

19 administration in vivo (Figure 3). Furthermore, this activation was completely confined to D1-

20 expressing neurons (i.e. >95% phospho-ERK positive neurons are D1-receptor/tdTomato

21 positive—see Figure 3). SKF81297-stimulated ERK phosphorylation was completely blocked in

22 the presence of 100 μM of EL1101 (Figure 3), indicating that the effects of D1 agonist treatment

15

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 on ERK activation are mediated directly at D1 dopaminoceptive medium spiny neurons (MSNs)

2 of the NAc, and are confined to these neurons.

3

4 Cocaine-induced up-regulation of ERK and Egr-1/Zif268 in mouse brain requires expression

5 of NCS-Rapgef2

6 We next sought to specifically manipulate Rapgef2 levels in dopaminoceptive brain

7 regions, the mPFC, BLA, and NAc, closely associated with behaviors affected by cocaine. NCS-

8 Rapgef2 knock-out specifically in corticolimbic areas, but not nucleus accumbens, was achieved

9 by crossing floxed Rapgef2 (Rapgef2cko/cko) with Camk2α-cre mice. NCS-Rapgef2 expression was

10 decreased at least 85% in mPFC and BLA in these mice, while NCS-

11 Rapgef2 expression in NAc was unaffected (Figure 4A). Knock-down of NCS-Rapgef2 expression

12 in D1 MSNs in NAc is not achievable by crossing a D1 receptor cre driver line with Rapgef2cko/cko

13 (Drd1-cre+/-:: Rapgef2cko/cko) (Extended Data Figure 4-1) or by crossing other striatum-specific Cre

14 driver lines, such as Gng7-Cre, with Rapgef2cko/cko mice (data not shown). Therefore, an alternative

15 method was employed to achieve the ablation of NCS-Rapgef2 expression in NAc, i.e. by bilateral

16 injection of AAV9-hSyn-Cre-eGFP into the ventral striatum of Rapgef2cko/cko mice (Figure 4B).

17 Expression of NCS-Rapgef2 in the NAc was not affected by injection of AAV9-hSyn-eGFP

18 control virus into Rapgef2cko/cko mice, or by injection of AAV9-hSyn-cre-eGFP into C57BL/6J

19 wild-type mice. However, NCS-Rapgef2 in NAc was significantly reduced by injection of AAV9-

20 hSyn-Cre-eGFP into Rapgef2cko/cko mice (Figure 4C).

21 Thus, NCS-Rapgef2 expression can be efficiently and specifically eliminated in NAc, by

22 stereotaxic AAC9-hSyn-Cre-eGFP injection in Rapgef2cko/cko mice, and in BLA and mPFC, by

23 genetic cross of Camk2α-cre+/- and Rapgef2cko/cko mice.

16

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Cocaine treatment is known to induce phospho-ERK elevation and trigger ERK-dependent

2 Egr-1/Zif268 expression in D1 medium-sized spiny neurons (Bertran-Gonzalez et al., 2008),

3 implicated in locomotor and drug preference responses to cocaine (Valjent et al., 2006c). To

4 confirm this, and to also confirm the existence of the ERKEgr-1/Zif268 pathway in D1-

5 dopaminoceptive neurons in mPFC and BLA, we treated mice in which D1 neurons are labeled

6 with tdTomato, with cocaine. Phospho-ERK neuronal induction after cocaine occurs

7 predominantly in D1-dopaminoceptive (tdTomato-positive) neurons in NAc, BLA and mPFC

8 (Figure 5A). We further quantified the percentage of pERK+ cells that were also D1 receptor-

9 expressing (tdTomato-positive) after treatment with cocaine. In prefrontal cortex, 56.7 ± 2.8% of

10 pERK+ neurons in layer II/III, 78.1 ± 2.4% of pERK+ neurons in layer V, and 97.3 ± 0.7% pERK+

11 neurons in layer VI were also tdTomato-positive. In BLA, 72.2 ± 2.8 % of pERK+ neurons were

12 D1 receptor positive, and in shell and core of NAc almost all (98.6 ± 0.1%) pERK+ neurons were

13 D1-MSNs.

14 We examined the requirement for NCS-Rapgef2 in cocaine-induced ERK

15 phosphorylation/activation, and subsequent Egr-1/Zif268 induction, in two sets of experiments.

16 First, the effects of cocaine injection were compared in Rapgef2cko/cko control mice, and in

17 Camk2α-cre+/-::Rapgef2cko/cko mice in which Rapgef2 expression is eliminated in mPFC and BLA

18 but not in NAc. ERK phosphorylation and Egr-1/Zif268 induction occurred in all three areas in

19 control mice at 10 and 60 min, respectively after i.p. cocaine administration (20 mg/kg). Induction

20 of both ERK phosphorylation and Egr-1/Zif268 was abolished in mPFC and BLA, where Rapgef2

21 expression was absent, but not in NAc, where Rapgef2 expression was spared (Figure 5B-D). Next,

22 the effects of cocaine injection were compared in Rapgef2cko/cko mice receiving AAV-Syn-Cre-

23 eGFP (to eliminate Rapgef2 expression in NAc) or AAV-Syn-eGFP (controls). ERK

17

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 phosphorylation and Egr-1/Zif268 induction by cocaine were elimninated in NAc after loss of

2 NCS-Rapgef2 expression in mice receiving intra-NAc AAV-Syn-Cre-eGFP injection (Figure 5E-

3 F) with no effect on ERK phosphorylation or Egr-1/Zif268 induction in mPFC or BLA (data not

4 shown). Similar results were obtained after ip injection of dopamine D1 receptor agonist

5 SKF81297 (Extended Data Figure 5-1). Thus, expression of NCS-Rapgef2 is required for cocaine-

6 induced up-regulation of ERK and Egr1/Zif268 in D1-expressing neurons of NAc, of mPFC, and

7 of BLA.

8 We note here that both D1 and D2 MSNs express NCS-Rapgef2 (Extended Data Figure 5-

9 2A). To determine if ERK activation in D2 MSNs requires NCS-Rapgef2, Rapgef2cko/cko mice

10 with NCS-Rapgef2 ablation in the NAc by AAV-Syn-cre-eGFP infusion were treated with the D2

11 antagonist eticlopride, which caused pERK induction in D2-MSN unaffected by ablation of NCS-

12 Rapgef2 expression (Extended Data Figure 5-2B).

13 .

14 NCS-Rapgef2 in D1 MSNs of ventral striatum, but not excitatory neurons in mPFC or BLA,

15 contributes to cocaine-induced psychomotor sensitization and conditioned place preference

16 Neither Camk2α-cre+/-::Rapgef2cko/cko mice, nor Rapgef2cko/cko mice infused with AAV-Syn-Cre-

17 eGFP in NAc, showed altered locomotor activity and anxiety in the elevated zero maze, indicated

18 by similar total distance traveled and time in open arms (Extended Data Figure 6-1), compared to

19 each other or to corresponding control mice. Mice were then subjected to testing for locomotor

20 sensitization to cocaine using the two-injection protocol (Valjent et al., 2010)(Figure 6A): For the

21 first 4 days, animals were habituated for 30 mins in an activity box, then injected with saline, and

22 allowed free activity for one hour. On day 5, after 30 min of habituation in the same box in the

23 same room, animals received their first dose of cocaine (15 mg/kg, ip). All four groups of animals

18

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 showed hyperactivity after cocaine injection (Figure 6B and 6C). On day 6, in the same context,

2 animals received a second dose of cocaine (15 mg/kg, ip) after the habituation trial. Both control

3 groups (Rapgef2cko/cko mice, and Rapgef2cko/cko mice injected with AAV-Syn-eGFP control virus

4 in the NAc) exhibited a comparable increase in locomotor activity compared with their response

5 to the first dose of cocaine (Figure 6B and 6C). Similar locomotor sensitization in response to

6 cocaine was observed in another control group: C57BL6/J wild-type mice injected with AAV-

7 Syn-cre-eGFP virus in the NAc (Extended Data Figure 6-2). Locomotor sensitization similarly

8 unaffected in Camk2α-cre+/-::Rapgef2cko/cko mice. However, cocaine-induced locomotor

9 sensitization was abolished in Rapgef2cko/cko mice with AAV-Syn-cre-eGFP bilateral infusion in

10 the NAc (Figure 6C).

11 We also studied the effects of region-specific NCS-Rapgef2 knockout on the effects of

12 repeated administration of cocaine on drug preference, using the conditioned place preference

13 (CPP) test (Figure 7A). Ablation of NCS-Rapgef2 in NAc by viral injection also abolished

14 cocaine-induced conditioned place preference (Figure 7B). These results indicate that two major

15 behavioral consequences of repeated psychomotor stimulant (cocaine) administration are

16 dependent on ERKEgr-1/Zif268 signaling in D1-MSNs, and further suggest cellular mechanistic

17 linkage between the two behaviors. CPP was not affected in Camk2α-cre+/-::Rapgef2cko/cko mice

18 (Figure 7C), indicating that the development of conditioned place preference inheres in cellular

19 changes in dopaminoceptive neurons of the nucleus accumbens, but not those in BLA and mPFC.

20

21 Parcellation of cAMP signaling to ERK and CREB in nucleus accumbens

22 Several models for intracellular signaling underlying dopamine-dependent ERK activation in D1

23 dopaminoceptive neurons emphasize that the sole cAMP effector for this process is PKA, both

19

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 because inhibition of ambient ERK phosphatase activity can be achieved through PKA-dependent

2 D1 signaling, and because previously no direct pathway for ERK activation by cAMP elevation

3 has been identified in neurons. To test whether or not the activation of ERK associated with

4 upregulation of Egr-1/Zif268 and locomotor sensitization/place preference conditioning by

5 cocaine involved activation of both PKA and NCS-Rapgef2, we quantified phospho-ERK and

6 phospho-CREB, as surrogates for NCS-Rapgef2 and PKA activation respectively, after cocaine

7 treatment in Rapgef2cko/cko control and NCS-Rapgef2-deficient mice. As shown in Figure 8, NCS-

8 Rapgef2 knockout in the NAc caused loss of ERK phosphorylation after cocaine treatment, while

9 cocaine-dependent CREB phosphorylation was preserved. Thus, PKA signaling is apparently

10 intact in mice in which both D1-MSN ERK activation, and acquisition of cocaine locomotor

11 sensitization and conditioned place preference, are lost after deletion of NCS-Rapgef2 in NAc.

20

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 DISCUSSION

2 Activation of ERK in vivo by D1 agonists. or the psychomotor stimulants cocaine and

3 amphetamine, in D1 neurons is well established (Valjent et al., 2000; Gutierrez-Arenas et al., 2014;

4 Jin et al., 2019). Expression of locomotor sensitization and CPP after cocaine administration is

5 ERK-dependent (Brami-Cherrier et al., 2005; Miller and Marshall, 2005; Ferguson et al., 2006;

6 Valjent et al., 2006b; Valjent et al., 2006a; Fasano et al., 2009), and is linked to ERK-dependent

7 transcription of the egr1 gene and expression of Egr1/Zif268 protein in dopaminoceptive neurons

8 in striatum and cerebral cortex (Valjent et al., 2006c; Gangarossa et al., 2011).

9 Whether dopamine signaling to ERK in D1 neurons is direct, or indirect via modulation

10 of ERK activation by other neurotransmitters including glutamate (Valjent et al., 2005) has been

11 an unresolved question. It has been hypothesized that glutamatergic activation of a ras-signaling

12 pathway for ERK phosphorylation is augmented by D1-dependent inhibition of ERK

13 dephosphorylation, shifting the balance between ERK activation/phosphorylation and

14 dephosphorylation towards phospho-ERK elevation and stimulation of ERK-dependent cellular

15 activation events, including egr1 gene transcription (Valjent et al., 2005; Lu et al., 2006; Santini

16 et al., 2007; Zhang et al., 2010; Santini et al., 2012). This mechanism has been favored because

17 striatal dopaminoceptive neurons possess a PKA- and dopamine responsive protein phosphatase

18 (DARPP-32) which disinhibits the striatally-enriched phosphatase STEP, leading to inhibition of

19 the putative proximate ERK phosphatase PP1 (Valjent et al., 2005; Sun et al., 2007; Gutierrez-

20 Arenas et al., 2014). However, differences between D1 signaling to ERK in ventral and dorsal

21 striatum (Gerfen et al., 2008) have occasioned a search for a direct intracellular signaling pathway

22 for ERK phosphorylation upon dopamine-stimulated elevation of intracellular cAMP. Indeed, G

23 protein-mediated cAMP pathway upregulation has been hypothesized as a common mechanism of

21

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 adaptation to chronic administration of drugs of abuse (Nestler, 2016), and ERK activation in

2 dopamine D1 receptor-expressing neurons in mesocorticolimbic projection areas, including both

3 medial prefrontal cortex and nucleus accumbens has been with drug-induced neuroadaptations

4 (Valjent et al., 2000; Adams and Sweatt, 2002; Valjent et al., 2005).

5 We have recently identified intracellular cAMP-dependent signaling downstream of Gs-

6 coupled receptors in neurons and endocrine cells via parallel and insulated (parcellated) pathways

7 initiated by the cAMP effectors PKA, Epac2, and NCS-Rapgef2. These effectors activate distinct

8 but coordinated cellular tasks. Thus, in neuroendocrine NS-1 cells, PKA signaling to CREB

9 mediates cell survival and neuron-specific gene expression; Epac2 signaling to the MAP kinase

10 p38 mediates growth arrest; and NCS-Rapgef2 signaling to the MAP kinase ERK mediates

11 neuritogenesis (Emery et al., 2013; Emery et al., 2014; Emery et al., 2017a). As the ERK pathway

12 is associated with neuritogenesis in NS-1 cells, and similar morphological changes occur via D1

13 signaling in NAc after cocaine (Lee et al., 2006), we hypothesized that the cAMPNCS-

14 Rapgef2ERK pathway might link D1 receptor activation in response to cocaine, to ERK

15 activation and downstream events involved in cellular plasticity causing behavioral effects of

16 repeated psychomotor stimulant administration.

17 The demonstration that NCS-Rapgef2 is required for Egr-1/Zif268 induction by cyclic

18 AMP in neuroendocrine cells, coupled with our previous finding of NCS-Rapgef2 dependence for

19 dopamine/D1 activation of neuritogenesis in these cells, suggests the existence of a direct

20 dopamineD1cAMPNCS-Rapgef2ERKEgr1/Zif268 signaling pathway with potential

21 relevance to cellular signaling in D1-MSNs in vivo. A role for NCS-Rapgef2 in mediating the

22 effects of increased synaptic dopamine after cocaine administration is further supported by the

23 localization of NCS-Rapgef2 to post-synaptic elements at dopaminergic synapses, and by NCS-

22

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Rapgef2-dependent activation of ERK by the D1 agonist SKF81297 in NAc brain slices ex vivo,

2 as reported here. Finally, we have studied the behavioral effects of cocaine in mice in which NCS-

3 Rapgef2 expression was eliminated in various brain regions (mPFC, BLA and NAc) in which

4 dopamine acts at D1 receptors. Loss of NCS-Rapgef2 expression in mPFC and BLA excitatory

5 neurons eliminated ERK activation by both SKF81297 and cocaine in these brain regions, without

6 affecting cocaine-induced locomotor sensitization, or conditioned place preference. Elimination

7 of NCS-Rapgef2 expression in NAc correspondingly abolished ERK activation by cocaine or D1

8 agonist, and also cocaine-induced locomotor sensitization and conditioned place preference. This

9 occurred without affecting cocaine-induced activation of CREB phosphorylation in MSNs. It

10 remains to be tested whether or not NCS-Rapgef2-dependent signaling in either mPFC or BLA is

11 required for cocaine-dependent behaviors not studied here, such as relapse (Rebec and Sun, 2005).

12 These findings have several implications for understanding the role of cyclic AMP in

13 mediating the post-synaptic actions of dopamine and subsequent dopamine-dependent behaviors,

14 and for defining the brain circuits in which dopamine signaling is critical for the various behavioral

15 consequences of acute and chronic cocaine ingestion.

16 Cocaine reinstatement depends on different cAMP-dependent pathways as a function of

17 the reinstatement stimulus: it is CREB-dependent for cocaine itself, and ERK-dependent for

18 restraint stress (Kreibich and Blendy, 2004; Briand and Blendy, 2013). These findings accord well

19 with our observation of parcellation of cAMP signaling to ERK and CREB through NCS-Rapgef2

20 and PKA, respectively, in cultured neuroendocrine cell lines (Emery et al., 2014). The present

21 report demonstrates that parcellation extends to D1-dependent activation of Egr-1/Zif268 via ERK

22 independently of CREB in NS-1 cells. Dopamine-dependent activation of ERK is likewise NCS-

23 Rapgef2-dependent, while signaling to CREB is not. This observation has allowed us to determine

23

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 that activation of ERK, apparently independently of PKA/CREB, and in dopaminoceptive neurons

2 of the NAc rather than mPFC or BLA, is required for the behavioral effects of repeated cocaine

3 administration.

4 ERK signaling itself is required for the sensitized locomotor, but not the initial locomotor

5 effects of cocaine administration (Valjent et al., 2006b). NCS-Rapgef2, however, may be an even

6 more practical target for mitigation of the neuroplastic effects of cocaine that underlie its addictive

7 potential, since it represents a single conditional input to ERK activation (see model, Figure 9). A

8 related member of the protein family present in brain, Rapgef6, mediates a different set of memory-

9 related functions in amygdala and hippocampus (Levy et al., 2015) and, interestingly, both

10 Rapgef2 and Rapgef6 are genetically linked to susceptibility to schizophrenia (Maeta et al., 2018).

11 The specialized behavioral functions of Rapgefs in the central nervous system, and the specific

12 pharmacological and behavioral effects of NCS-Rapgef2 demonstrated here, suggests that NCS-

13 Rapgef2 merits consideration as a target for other reward-related psychopathology besides

14 addiction.

15 Convergent activation of ERK activation in D1-MSNs through direct (NCS-Rapgef2-

16 mediated) and indirect (PKA-mediated) dopaminergic signaling, and via glutamatergic signaling,

17 provides a mechanistically complete picture of D1-MSN cellular plasticity during repeated

18 exposure to psychomotor stimulants. It has implications for understanding, and intervening in,

19 psychomotor stimulant addiction. In particular, the model (Figure 9) implies that while blockade

20 of D1 signaling altogether could lead to abolition of cocaine-induced locomotor activation, as

21 occurs in D1 receptor deficient (Xu et al., 1994) or D1-MSN ERK-deleted mice (Brami-Cherrier

22 et al., 2005; Ferguson et al., 2006), inhibition of either the NCS-Rapgef2 or PKA parcellated

24

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 signaling pathways may lead to selective blockade of different subsets of learned behaviors

2 occurring during chronic cocaine administration.

3 Cocaine administration causes ERK activation predominantly if not exclusively in D1-

4 compared to D2-MSNs in the ventral striatum (Valjent et al., 2005; Bertran-Gonzalez et al., 2008),

5 and D1 receptor-expressing neurons play a predominant role in cocaine sensitization and reward

6 learning (see (Baik, 2013) and references therein). Recent studies using optogenetic manipulation

7 and in vivo calcium imaging have confirmed the view that activation of D1 neurons promotes the

8 formation of cocaine reward (Lobo et al., 2010; Kravitz et al., 2012; Chandra et al., 2013; Calipari

9 et al., 2016). NCS-Rapgef2 is expressed in both D1 and D2 neurons (Extended Data Figure 5-1).

10 While ERK activation does not occur in D2-MSNs after cocaine administration, it is activated in

11 in this neuronal population by the D2 receptor antagonist eticlopride. However, this response was

12 not impaired in NCS-Rapgef2-deficient mice (Extended Data figure 5-2). Thus, the NCS-Rapgef2-

13 dependent activation of ERK in D1-MSN signaling documented here is likely to be the major

14 action of this signaling molecule in mediating the cocaine-dependent behaviors examined here.

15 D1- and D2-MSNs in NAc can drive both reward and aversion, depending on their stimulation

16 pattern (Soares-Cunha et al., 2019): it remains to be determined whether or not NCS-Rapgef2

17 signaling in D2 neurons contributes to these other aspects of NAc function.

18 Additional questions remain: what is the role of the NCS-Rapgef2 pathway in D1 signaling

19 outside of the NAc? Might signaling through NCS-Rapgef2 mediate cocaine relapse associated

20 with dopamine neurotransmission in mPFC (Miller and Marshall, 2005; Lu et al., 2006; Girault et

21 al., 2007)? What might be the role(s) of CREB-dependent signaling (Shaywitz and Greenberg,

22 1999), spared upon abrogation of NCS-Rapgef2, in the D1-MSN response to cocaine? Finally,

23 might it be possible to develop brain-permeant NCS-Rapgef2 antagonists of sufficient potency and

25

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 selectivity for use in vivo? If dendritic spinogenesis in D1-MSNs is the morphological correlate of

2 Rapgef2-dependent ERK signaling to Egr-1/Zif268 resulting in neuritogenesis in NS-1 cells, it

3 might be supposed that blocking this process could affect not only short-term effects of repeated

4 cocaine administration including locomotor sensitization and cocaine preference, but also longer-

5 term effects of cocaine-seeking behavior associated with altered D1-MSN spine density (Lee et

6 al., 2006; Ren et al., 2010).

7

26

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 FIGURE LEGENDS

2 Figure 1 NCS-Rapgef2 dependent phospho-ERK activation and Egr-1 upregulation in

3 neuroendocrine cell line NS-1 induced by 8-CPT-cAMP. (A) NS-1 cells (NS-1(Rapgef2 +/+)) or

4 NS-1 cells with NCS-Rapgef2 KO by CRISPR (NS-1 (Rapgef2 -/-) ) were treated with 100 µM

5 8-CPT-cAMP or 0.01% DMSO (8-CPT-cAMP (-) group) for 1 hour, then the cell lysate were

6 collected for western blot analysis of pan-ERK, phospho-ERK and Egr-1. Equal amount of total

7 protein (20 µg) was loaded in each lane. (B) Phospho-ERK and Egr-1 protein levels were

8 quantified with Image J and compared to NS1 (Rapgef2+/+)/8-CPT-cAMP (-) group (n=3 per

9 group). post hoc Bonferroni test following two-way ANOVA, *p<0.05. **p<0.001.

10

11 Figure 2 Postsynaptic localization of NCS-Rapgef2 in dopaminoceptive neurons in mPFC, NAc

12 and BLA. (A) NCS-Rapgef2 immunoreactive (IR) signal (red) is not colocalized with Bassoon

13 (green), a scaffolding protein of presynaptic active zone, but in close apposition to the Bassoon

14 puncta, indicated by white arrows in a1 & a2 (mPFC), b1 & b2 (NAc) and c1 & c2 (BLA). Scale

15 bar: 20 µm (left); 5 µm (middle); 1 µm (right). (B) NCS-Rapgef2 IR signal (red) is colocalized

16 with postsynaptic marker PSD95 IR signal (green) in the soma and dendrites (indicated by white

17 arrows) of neurons in mPFC pyramidal cells, NAc medium spiny neurons and excitatory neurons

18 in BLA. Right panels show, at higher magnification, the images outlined by white boxes on the

19 left. Scale bar: 20 µm (left); 5 µm (right).

20

21 Figure 3 NCS-Rapgef2 inhibitor N6-phenyl, 9-tetrahydrofuranyl adenine (EL1101)

22 attenuated phospho-ERK activation in the NAc of mouse brain slice induced by D1 receptor

23 agonist SKF 81297.

27

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 (A) Brain slices (coronal sections in 250 µm) were incubated vehicle (DMSO final concentration

2 0.1%), 10 µM SKF81297, 10 µM U0126 plus 10 µM SKF81297 or 100 µΜ EL1101 plus 10

3 µΜ SKF81297 for 30 min. After fixation with 4% PFA, slices were stained with phospho-ERK

4 antibody. Phospho-ERK activation in the NAc of mouse brain slice can be induced by D1 receptor

5 agonist SKF81297, that was blocked by the MEK1 inhibitor U0126 or attenuated by NCS-Rapgef2

6 inhibitor EL1011. Right panels show, at higher magnification, the images outlined by white boxes

7 on the left. Scale bar: 500 µm (left); 100 µm (right). (B) D1-specific activation of phospho-ERK

8 in the NAc of D1d1a-tdTomato mouse indicated by colocalization of phospho-ERK IR signal

9 (green) and D1 MSNs (tdTomato, red). Scale bar: 50 µm (left).

10

11 Figure 4 Region-specific ablation of NCS-Rapgef2 expression.

12 (A) Camk2α-cre+/-::Rapgef2cko/cko (cKO) was generated by crossing Rapgef2cko/cko (flox) with

13 Camk2α-cre mice. NCS-Rapgef2 expression was largely ablated in mPFC and BLA of cKO, while

14 was unaffected in NAc of cKO. Scale bar: 100 µm in mPFC and BLA images; 50 µm & 10 µm in

15 NAc. N=4~7 animals per group. Student’s t-test for each region, *p<0.001.

16 (B) Representative image of brain slice 4 weeks after AAV bilateral viral injection in NAc.

17 (C) NCS-Rapgef2 in NAc was abrogated by bilateral injection of AAV9-hSyn-cre-eGFP (cre

18 virus) into the ventral striatum of Rapgef2cko/cko (flox) mice. However, expression of NCS-Rapgef2

19 in the NAc was not affected by injection of AAV9-hSyn-eGFP (ctrl virus) into Rapgef2cko/cko (flox)

20 mice or by injection of AAV9-hSyn-cre-eGFP (cre virus) into C57BL/6J wild-type mice. Bottom

21 panels show, at higher magnification, the images outlined by white boxes in upper panels. Scale

22 bar: 20 µm (top) and 10 µm (bottom). NCS-Rapgef2 IR signals in GFP-positive neurons were

28

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 quantified with ImageJ. N=4~7 animals per group. post hoc Bonferroni test following one-way

2 ANOVA, *p<0.05.

3

4 Figure 5 Expression of NCS-Rapgef2 is required for cocaine-induced up-regulation of ERK and

5 Egr1/zif268 in mouse brain

6 (A) Majority of cocaine-induced phospho-ERK activation (10 min after administration of 20

7 mg/kg cocaine, ip) was in D1 receptor positive neurons in mPFC, NAc and BLA. Right panels

8 (indicated as a, b, c) show the images at higher magnification, outlined by white boxes on the left.

9 Scale bar: 200 µm.

10 (B) Phospho-ERK elevation observed 10 mins after ip administration of 20 mg/kg cocaine in

11 Rapgef2cko/cko (flox) mice, is attenuated in mPFC and BLA, but not in NAc, of Camk2α-cre+/-

12 ::Rapgef2cko/cko (cKO) mice. Scale bar: 200 µm.

13 (C) Egr-1/zif268 elevation observed 1 h after 20 mg/kg cocaine treatment in Rapgef2cko/cko (flox)

14 mice is attenuated in mPFC and BLA, but not in NAc, of Camk2α-cre+/-::Rapgef2cko/cko (cKO)

15 mice. Scale bar: 100 µm.

16 (D) Quantification of phospho-ERK and Egr-1/zif268 in Rapgef2cko/cko (flox) or Camk2α-cre+/-

17 ::Rapgef2cko/cko (cKO) mice after saline or cocaine treatment. Relative number of phospho-ERK

18 positive neurons or Egr-1 IR from mPFC, NAc and BLA of each animal was normalized by the

19 average value from age-matched saline-treated Rapgef2cko/cko (flox) mice. N=3~6 animals per

20 group. post hoc Bonferroni test following two-way ANOVA, *p < 0.05. (phospho-ERK in mPFC:

21 genotype effect F (1, 13) = 21.312, p<0.001, drug effect F (1, 13) = 43.108, p<0.001, interaction F (1,

22 13) = 21.502, p<0.001; phospho-ERK in NAc, genotype effect F (1, 10) = 2.131, p=0.175, drug effect

23 F (1, 10) = 433.739, p<0.001, interaction F (1, 10) = 2.427, p=0.15; phospho-ERK in BLA: genotype

29

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 effect F (1, 16) = 14.395, p=0.002, drug effect F (1, 16) = 26.668, p<0.001, interaction F (1, 16) = 23.691,

2 p<0.001; Egr-1 in mPFC: genotype effect F (1, 15) = 10.348, p=0.006, drug effect F (1, 15) = 11.435,

3 p=0.004, interaction F (1, 15) = 11.62, p=0.004; Egr-1 in NAc: genotype effect F (1, 16) = 0.00125,

4 p=0.972, drug effect F (1, 16) = 169.616, p<0.001, interaction F (1, 16) = 0.11, p=0.744; Egr-1 in BLA:

5 genotype effect F (1, 13) = 19.872, p<0.001, drug effect F (1, 13) = 15.786, p=0.002, interaction F (1,

6 13) = 29.799, p<0.001)

7 (E) Bilateral ablation of NCS-Rapgef2 in NAc impaired phospho-ERK elevation in response to

8 cocaine. AAV9-hSyn-eGFP (ctrl virus) or AAV9-hSyn-cre-eGFP (cre virus) was injected into the

9 NAc of Rapgef2cko/cko mice. Four weeks later, phospho-ERK activation in both sides of NAc was

10 examined by immunohistochemistry after acute cocaine (ip, 20 mg/kg, 10 min) injection. N=3~4

11 animals per group. post hoc Bonferroni test following two-way ANOVA, *p < 0.05. (phospho-

12 ERK in NAc: genotype effect F (1, 9) = 12.262, p=0.007, drug effect F (1, 9) = 13.901, p=0.005,

13 interaction F (1, 9) = 18.216, p=0.002)

14 (F) Cocaine-induced Egr-1/zif268 increase in the NAc of Rapgef2cko/cko mice with ctrl virus, was

15 impaired in the NAc with cre virus. Four weeks after viral infusion, mice were sacrificed 1 hr after

16 saline or cocaine (20 mg/kg) injection. The tissue from NAc was punched out from 0.5 mm coronal

17 sections of mouse brains and protein lysate was used for western blot with Egr-1/zif268 antibody.

18 Total protein loading of each sample was first normalized by GAPDH immunoreactivity (IR).

19 Relative Egr-1/zif268 IR (means ± SEM) was obtained by comparing to average of that from NAc

20 of saline-treated Rapgef2cko/cko bilaterally injected with AAV9-hSyn-eGFP (ctrl virus). N=4~5

21 animals per group. post hoc Bonferroni test following two-way ANOVA, *p < 0.05. (Egr-1 in

22 NAc: genotype effect F (1, 15) =31.507, p<0.001; drug effect F (1, 15) =7.995, p=0.013; genotype X

23 drug effect F (1, 15) =21.792, p<0.001)

30

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1

2 Figure 6. Locomotor sensitization induced by cocaine was abolished in the mice with NCS-

3 Rapgef2 ablation in the NAc.

4 (A) Experimental design for locomotor sensitization. Camk2α-cre+/-::Rapgef2cko/cko (cKO) mice,

5 Rapgef2cko/cko (flox) mice with bilateral NAc AAV9-hSyn-cre-eGFP (cre virus) injection, and their

6 corresponding controls were subjected to a two-injection protocol (TIPS) for cocaine locomotor

7 sensitization. For the first 4 days, animals were habituated for 30 min in an activity chamber, then

8 injected with saline, and allowed free movement within the chamber for 60 min. On day 5, after

9 30 min of habituation in the same box in the same room, animals received the first dose of cocaine.

10 On day 6, in the same context (same box in same room), animals received a second dose of cocaine

11 after a habituation trial.

12 (B) Animal locomotor activity in response to saline or cocaine administration. Activity was

13 monitored in two trials each day: 30 minutes free running and 1 hour after saline or cocaine (15

14 mg/kg) injection.

15 (C) Locomotor activity (total number of beam breaks) for one hour following saline or cocaine

16 administration. SAL Avg, average of activity during 4 days of saline injection; COC1, activity

17 after the first dose of cocaine (15 mg/kg) on day 5; COC2, activity after the second dose of cocaine

18 (15 mg/kg) on day 6. N=12 animals per group. Two-way ANOVA followed by post hoc Fisher’s

19 LSD test, *p<0.05 ** p<0.001. (genotype effect: F (3, 129) =1.804, p=0.15; drug effect: F (2, 129)

20 =73.632, p<0.001; interaction: F (6, 129) =1.201, p=0.31. No difference between groups within SAL

21 Avg; No difference between groups within COC1; cre virus group is different from either group

22 within COC2).

31

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 (D) Schematic representation of the injection sites in the NAc for the mice used for locomotor

2 sensitization test. N=12 animals for control virus injection; n=12 animals for cre virus injection.

3

4 Figure 7 Conditioned place preference (CPP) induced by cocaine was impaired in the mice with

5 NCS-Rapgef2 KO in the NAc.

6 (A) Experimental design for CPP. A shuttle box with a door that can be opened to allow the animal

7 free access to the two chambers was used for CPP. The two chambers differed on several sensory

8 and environmental properties. Animals received 8 days /four-sets of cocaine and saline-pairing:

9 for cocaine group, pair one chamber with cocaine, pair opposite one with saline, for saline group,

10 pair both sides with saline. Animals were tested for the place preference before drug pairing and

11 postconditioning by recording the amount of time they spent in each chamber.

12 (B) CPP scores for mice with NCS-Rapgef2 KO in the NAc compared to the corresponding

13 controls. N=7~10 animals per group. post hoc Bonferroni test following three-way ANOVA,

14 *p<0.05. (pre- vs post-conditioning: F (1, 58) =36.937, p<0.001; genotype effect: F (1, 58) =3.059,

15 p=0.086; drug effect: F (1, 58) =5.687, p=0.02; conditioning X genotype interaction: F (1, 58) =4.875,

16 p=0.031; conditioning X drug interaction: F (1, 58) =4.5, p=0.038; conditioning X genotype X drug

17 interaction: F (1, 58) =0.443, p=0.508).

18 (C) CPP scores for Camk2α-cre+/-::Rapgef2cko/cko (cKO) mice compared to the corresponding

19 controls Rapgef2cko/cko (flox). N=10~16 animals per group. Post hoc Bonferroni test following

20 three-way ANOVA, *p<0.05. **p<0.001. (pre- vs post-conditioning: F (1, 90) =67.764, p<0.001;

21 genotype effect: F (1, 90) =0.315, p=0.576; drug effect: F (1, 90) =11.629, p<0.001; conditioning X

22 genotype interaction: F (1, 90) =0.969, p=0.328; conditioning X drug interaction: F (1, 90) =12.82,

32

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 p<0.001; genotype X drug interaction: F (1, 90) =0.0099, p=0.921; conditioning X genotype X drug

2 interaction: F (1, 90) =5.332, p=0.023)

3 (D) Schematic representation of the injection sites in the NAc for the mice used for CPP test. N=18

4 animals for ctrl virus injection: n=8 animals for saline group, n=10 animals for cocaine group.

5 N=15 animals for cre virus injection: n=8 animals for saline group, n=7 animals for cocaine group.

6

7 Figure 8. Cocaine treatment resulted in ERK phosphorylation in an NCS-Rapgef2 dependent

8 manner, while CREB phosphorylation was unaffected by NCS-Rapgef2 ablation. AAV9-hSyn-

9 eGFP (ctrl virus) or AAV9-hSyn-cre-eGFP (cre virus) was injected into both sides of NAc of

10 Rapgef2cko/cko mice. After four weeks, saline or cocaine (20 mg/kg) was injected (ip) and animals

11 were perfused 10 min later for phospho-ERK and phospho-CREB immunohistochemistry. The

12 number of phospho-ERK/GPF co-positive neurons was significantly increased in the NAc with

13 ctrl virus after cocaine treatment, but not in the NAc with cre virus (n= 3~4 animals per group;

14 genotype effect F(1, 9) = 7.84, p=0.021; drug effect F(1, 9)=7.716, p=0.021; genotype X drug effect

15 F (1, 9) =11.374, p=0.008, two-way ANOVA). Phospho-CREB elevation induced by cocaine

16 treatment was not affected by NCS-Rapgef2 ablation in the NAc (n= 3~4 animals per group;

17 genotype effect F(1, 9) = 0.173, p=0.687; drug effect F(1, 9)=84.582, p<0.001; genotype X drug effect

18 F (1, 9) =0.058, p=0.815, two-way ANOVA). Two-way ANOVA followed by post hoc Bonferroni

19 test, *p < 0.05, **<0.001. Scale bars: 50 µm.

20

21 Figure 9. Proposed direct model for dopamine-dependent ERK activation in D1-

22 dopaminoceptive neurons.

33

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Model for convergent ERK activation by D1 receptor activation adapted from summary of

2 previous conceptualization of this signaling network by Pascoli et al. (Pascoli et al., 2014).

3 Psychomotor stimulants such as cocaine act by increasing synaptic dopamine leading to ERK

4 activation in nucleus accumbens which is required for long-term behavioral adaptations such as

5 locomotor sensitization and CPP. It has previously been thought that dopamine acting through the

6 D1 receptor affects ERK activity only indirectly, via PKA- and DARPP-32/STEP-mediated

7 inhibition of ERK dephosphorylation (Svenningsson et al., 2004; Valjent et al., 2005), while direct

8 ERK activation itself occurs in the D1 medium spiny neuron only via NMDA receptor-dependent

9 glutamatergic signaling (Pascoli et al., 2011; Cahill et al., 2014). Calcium influx via NMDARs

10 activates calcium-sensitive Ras-guanine nucleotide releasing factor (Ras-GRF1) that in turn can

11 activate the Ras-Raf-MEK cascade (Farnsworth et al., 1995; Fasano et al., 2009). According to

12 this model, the summation of ERK activation by glutamate and of phospho-ERK stabilization by

13 dopamine activity leads to activity-dependent ERK translocation to the nucleus, where it controls

14 both chromatin remodeling and gene transcription, with one of its targets being MSK1/CREB.

15 This model for dopamine signaling in D1-MSNs requires revision (see discussion). First Nagai

16 and colleagues have demonstrated that dopamine can stimulate Rap1 GEF (Rasgrp2)

17 phosphorylation via PKA in MSNs of ventral striatum, implying that ERK could be activated

18 directly by dopamine through Rap1 (Nagai et al., 2016b; Nagai et al., 2016a). We propose here a

19 more parsimonious mechanism, and one more directly supported by experimental evidence, for

20 direct activation of ERK by D1-dependent dopamine signaling, via cAMP activation of NCS-

21 Rapgef2. Thus, we demonstrate here that NCS-Rapgef2 elimination from MSNs in NAc abolishes

22 dopamine-D1-cAMP-dependent activation of ERK, while dopamine-D1-cAMP-PKA-dependent

23 activation of CREB proceeds intact. Thus, cAMP elevation by dopamine in D1-MSNs results not

34

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 in activation of ERK and CREB in series, but as a parcellation of signaling between ERK and

2 CREB resulting in separate cellular consequences under the control of each pathway. We note that

3 our finding of a direct pathway for D1 activation of ERK through NCS-Rapgef2 does not

4 contradict evidence for an indirect modulatory role for PKA-dependent ERK phosphatase

5 inhibition after psychomotor stimulant administration (Valjent et al., 2005; Sun et al., 2007), and

6 is also not inconsistent with additional ERK regulation by glutamatergic input to D1

7 dopaminoceptive neurons in the context of cellular plasticity underlying cocaine addiction (Valjent

8 et al., 2000; Park et al., 2013; Cahill et al., 2014; Pascoli et al., 2014). However, it may explain the

9 relative mild, yet more pleiotropic effect(s) of DARPP-32 knock-out (Svenningsson et al., 2004),

10 while the effects of NCS-Rapgef2 are both more marked, and more specific. We posit that D1

11 receptor activation, and cAMP elevation, in D1 MSNs likely results in parallel effects on ERK,

12 both directly via NCS-Rapgef2 and indirectly via PKA. In this model, the effects of cocaine rely

13 on multiple necessary, but perhaps individually insufficient inputs activated by dopamine, that

14 converge on D1-MSN ERK phosphorylation. These inputs include dopamine action through cyclic

15 AMP and PKA/DARPP/STEP/PP1 (Svenningsson et al., 2004), through cyclic AMP and

16 PKA/RasGRP2/Rap1 (Nagai et al., 2016b), a src-dependent non-cAMP-mediated augmentation of

17 NMDA receptor-dependent ERK activation via ras (Fasano et al., 2009; Pascoli et al., 2011), and

18 a direct pathway to ERK activation by D1 receptor activation via cyclic AMP and NCS-

19 Rapgef2/Rap1/B-Raf/MEK.

20

21 Extended Data Figure 4-1. Cre expression in D1d1a-cre mouse line is not strong enough to

22 knockout NCS-Rapgef2 in D1 MSNs in NAc.

35

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 (A) NCS-Rapgef2 expression is largely intact in the NAc (top), especially in D1-MSNs (bottom,

2 in green) of Drd1-cre+/-:: Rosa-eGFP:: Rapgef2cko/cko compared to Drd1-cre+/-::Rosa-eGFP,

3 indicated by Rapgef2 antibody staining. (B) Phospho-ERK activation in the NAc of Drd1-cre+/-::

4 Rapgef2cko/cko was normally induced by D1 receptor agonist SKF81297 (ip, 2 mg/kg, 15 min).

5

6 Extended Data Figure 5-1. Phospho-ERK elevation observed 30 mins after ip administration of

7 5 mg/kg SKF81927 in Rapgef2cko/cko (flox) mice, is attenuated in mPFC and BLA, but not in NAc,

8 of Camk2α-cre+/-::Rapgef2cko/cko (cKO) mice. Scale bar: 200 µm.

9

10 Extended Data Figure 5-2. (A) NCS-Rapgef2 is expressed in both D1 and D2 MSNs in NAc.

11 NCS-Rapgef2 immunohistochemistry was performed using brain sections from Drd1-cre+/-::Rosa-

12 eGFP or Drd2-cre+/-::Rosa-eGFP mice. NCS-Rapgef2 IR signal (red) was seen in either D1 MSNs,

13 indicated by GFP IR signal (green) in the NAc of Drd1-cre+/-::Rosa-eGFP mice, or in D2 MSNs

14 indicated by GFP IR signal (green) in the NAc of Drd2-cre+/-::Rosa-eGFP mice. (B) pERK

15 induction in presumptive D2-MSNs in NAc after in vivo eticlopride administration is not NCS-

16 Rapgef2-dependent. Phospho-ERK activation in NAc was measured in NAc of Rapgef2cko/cko mice

17 in which either AAV9-hSyn-eGFP (ctrl virus) or AAV9-hSyn-cre-eGFP (cre virus) was injected

18 into NAc, and four weeks later mice were treated with either saline or the D2R antagonist

19 eticlopride (2 mg/kg, ip, 15 min). Total number of phospho-ERK positive neurons and phospho-

20 ERK positive neurons with GFP positive (pERK+ & GFP+) in NAc were quantified with ImageJ.

21 Relative number of phospho-ERK positive neurons in NAc of each animal was normalized by the

22 average value from saline-treated Rapgef2cko/cko (flox) mice with ctrl virus. N=3 animals per group.

23 post hoc Bonferroni test following two-way ANOVA, *p < 0.05. Scale bars: 50 µm.

36

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1

2 Extended Data Figure 6-1. Neither Camk2α-cre+/-::Rapgef2cko/cko (cKO) mice nor Rapgef2cko/cko

3 (flox) mice with AAV9-hSyn-cre-eGFP (cre virus) injection in the NAc showed changes in anxiety

4 and locomotor activity, compared to their corresponding controls, in the zero maze test. All

5 animals were housed in divided cages before behavioral experiments.

6

7 Extended Data Figure 6-2. Locomotor sensitization can be induced by cocaine in C57BL/6 wild-

8 type mice with cre viral infusion in the NAc bilaterally. Same two-injection protocol as figure 6

9 was used. C57BL/6N (n=13) or C57BL/6J (n=12) were injected with AAV9-hSyn-cre-eGFP

10 bilaterally (0.3 μl of virus/each side, ~0.5x109 infectious particle per microliter). Four weeks later,

11 animals were subjected to locomotor sensitization task. C57BL/6J (panel B) showed much stronger

12 response to cocaine than C57BL/6N (panel A). (C) Locomotor activity (total number of beam

13 breaks) for one hour following saline or cocaine administration. C57BL/6J with cre virus injection

14 in the NAc showed similar locomotor sensitization in response to cocaine (15 mg/kg) as

15 Rapgef2cko/cko mice or Rapgef2cko/cko injected with control virus indicated in figure 6. Two-way

16 ANOVA followed by post hoc Fisher’s LSD test, *p<0.05. (C57BL/6J vs C57BL/6N: F (1, 69)

17 =34.928, p<0.001; SAL Avg vs COC1 vs COC2: F (2, 69) =27.006, p<0.001; genotype X drug

18 interaction: F (2, 69) =1.337, p=0.269.).

19

20

37

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 REFERENCES

2 Adams JP, Sweatt JD (2002) Molecular psychology: roles for the ERK MAP kinase cascade in

3 memory. Annu Rev Pharmacol Toxicol 42:135-163.

4 Baik JH (2013) Dopamine signaling in reward-related behaviors. Frontiers in neural circuits 7:152.

5 Bertran-Gonzalez J, Bosch C, Maroteaux M, Matamales M, Herve D, Valjent E, Girault JA (2008)

6 Opposing patterns of signaling activation in dopamine D1 and D2 receptor-expressing

7 striatal neurons in response to cocaine and haloperidol. J Neurosci 28:5671-5685.

8 Brami-Cherrier K, Valjent E, Herve D, Darragh J, Corvol JC, Pages C, Arthur SJ, Girault JA,

9 Caboche J (2005) Parsing molecular and behavioral effects of cocaine in mitogen- and

10 stress-activated protein kinase-1-deficient mice. J Neurosci 25:11444-11454.

11 Briand LA, Blendy JA (2013) Not all stress is equal: CREB is not necessary for restraint stress

12 reinstatement of cocaine-conditioned reward. Behav Brain Res 246:63-68.

13 Cahill E, Salery M, Vanhoutte P, Caboche J (2014) Convergence of dopamine and glutamate

14 signaling onto striatal ERK activation in response to drugs of abuse. Frontiers in

15 pharmacology 4:172.

16 Calipari ES, Bagot RC, Purushothaman I, Davidson TJ, Yorgason JT, Pena CJ, Walker DM,

17 Pirpinias ST, Guise KG, Ramakrishnan C, Deisseroth K, Nestler EJ (2016) In vivo imaging

18 identifies temporal signature of D1 and D2 medium spiny neurons in cocaine reward. Proc

19 Natl Acad Sci U S A 113:2726-2731.

20 Chandra R, Lenz JD, Gancarz AM, Chaudhury D, Schroeder GL, Han MH, Cheer JF, Dietz DM,

21 Lobo MK (2013) Optogenetic inhibition of D1R containing nucleus accumbens neurons

22 alters cocaine-mediated regulation of Tiam1. Front Mol Neurosci 6:13.

38

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Emery A, Eiden MV, Mustafa T, Eiden LE (2013) GPCR-Gs signaling to ERK is controlled by

2 the cAMP-sensing guanine nucleotide exchange factor NCS/Rapgef2 in neuronal and

3 endocrine cells. Science Signaling 6:ra51.

4 Emery AC, Eiden LE (2012) Signaling through the neuropeptide GPCR PAC1 induces

5 neuritogenesis via a single linear cAMP- and ERK-dependent pathway using a novel

6 cAMP sensor. FASEB J 26:3199-3211.

7 Emery AC, Eiden MV, Eiden LE (2014) Separate cyclic AMP sensors for neuritogenesis, growth

8 arrest, and survival of neuroendocrine cells. J Biol Chem 289:10126-10139.

9 Emery AC, Xu W, Eiden MV, Eiden LE (2017a) Guanine nucleotide exchange factor Epac2-

10 dependent activation of the GTP-binding protein Rap2A mediates cAMP-dependent

11 growth arrest in neuroendocrine cells. J Biol Chem 292:12220-12231.

12 Emery AC, Alvarez RA, Eiden MV, Xu W, Simeon FG, Eiden LE (2017b) Differential

13 Pharmacophore Definition of the cAMP Binding Sites of Neuritogenic cAMP Sensor-

14 Rapgef2, Protein Kinase A, and Exchange Protein Activated by cAMP in Neuroendocrine

15 Cells Using an Adenine-Based Scaffold. ACS Chem Neurosci 8:1500-1509.

16 Farnsworth CL, Freshney NW, Rosen LB, Ghosh A, Greenberg ME, Feig LA (1995) Calcium

17 activation of Ras mediated by neuronal exchange factor Ras-GRF. Nature 376:524-527.

18 Fasano S, D'Antoni A, Orban PC, Valjent E, Putignano E, Vara H, Pizzorusso T, Giustetto M,

19 Yoon B, Soloway P, Maldonado R, Caboche J, Brambilla R (2009) Ras-guanine

20 nucleotide-releasing factor 1 (Ras-GRF1) controls activation of extracellular signal-

21 regulated kinase (ERK) signaling in the striatum and long-term behavioral responses to

22 cocaine. Biol Psychiatry 66:758-768.

39

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Ferguson SM, Fasano S, Yang P, Brambilla R, Robinson TE (2006) Knockout of ERK1 enhances

2 cocaine-evoked immediate early gene expression and behavioral plasticity.

3 Neuropsychopharmacology 31:2660-2668.

4 Fernando AB, Robbins TW (2011) Animal models of neuropsychiatric disorders. Annu Rev Clin

5 Psychol 7:39-61.

6 Funahashi Y, Ariza A, Emi R, Xu Y, Shan W, Suzuki K, Kozawa S, Ahammad RU, Wu M, Takano

7 T, Yura Y, Kuroda K, Nagai T, Amano M, Yamada K, Kaibuchi K (2019) Phosphorylation

8 of Npas4 by MAPK Regulates Reward-Related Gene Expression and Behaviors. Cell

9 reports 29:3235-3252 e3239.

10 Gangarossa G, Di Benedetto M, O'Sullivan GJ, Dunleavy M, Alcacer C, Bonito-Oliva A, Henshall

11 DC, Waddington JL, Valjent E, Fisone G (2011) Convulsant doses of a dopamine D1

12 receptor agonist result in Erk-dependent increases in Zif268 and Arc/Arg3.1 expression in

13 mouse dentate gyrus. PLoS One 6:e19415.

14 Gerfen CR, Paletzki R, Worley P (2008) Differences between dorsal and ventral striatum in Drd1a

15 dopamine receptor coupling of dopamine- and cAMP-regulated phosphoprotein-32 to

16 activation of extracellular signal-regulated kinase. J Neurosci 28:7113-7120.

17 Girault JA, Valjent E, Caboche J, Herve D (2007) ERK2: a logical AND gate critical for drug-

18 induced plasticity? Curr Opin Pharmacol 7:77-85.

19 Gutierrez-Arenas O, Eriksson O, Kotaleski JH (2014) Segregation and crosstalk of D1 receptor-

20 mediated activation of ERK in striatal medium spiny neurons upon acute administration of

21 psychostimulants. PLoS Comput Biol 10:e1003445.

40

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Heien ML, Khan AS, Ariansen JL, Cheer JF, Phillips PE, Wassum KM, Wightman RM (2005)

2 Real-time measurement of dopamine fluctuations after cocaine in the brain of behaving

3 rats. Proc Natl Acad Sci U S A 102:10023-10028.

4 Hikida T, Kimura K, Wada N, Funabiki K, Nakanishi S (2010) Distinct roles of synaptic

5 transmission in direct and indirect striatal pathways to reward and aversive behavior.

6 Neuron 66:896-907.

7 Hutton SR, Otis JM, Kim EM, Lamsal Y, Stuber GD, Snider WD (2017) ERK/MAPK Signaling

8 Is Required for Pathway-Specific Striatal Motor Functions. J Neurosci 37:8102-8115.

9 Jiang SZ, Xu W, Emery AC, Gerfen CR, Eiden MV, Eiden LE (2017) NCS-Rapgef2, the Protein

10 Product of the Neuronal Rapgef2 Gene, Is a Specific Activator of D1 Dopamine Receptor-

11 Dependent ERK Phosphorylation in Mouse Brain. eNeuro 4.

12 Jin DZ, Mao LM, Wang JQ (2019) Amphetamine activates non-receptor tyrosine kinase Fyn and

13 stimulates ERK phosphorylation in the rat striatum in vivo. Eur J Pharmacol 843:45-54.

14 Kravitz AV, Tye LD, Kreitzer AC (2012) Distinct roles for direct and indirect pathway striatal

15 neurons in reinforcement. Nat Neurosci 15:816-818.

16 Kreibich AS, Blendy JA (2004) cAMP response element-binding protein is required for stress but

17 not cocaine-induced reinstatement. J Neurosci 24:6686-6692.

18 Laakso A, Mohn AR, Gainetdinov RR, Caron MG (2002) Experimental genetic approaches to

19 addiction. Neuron 36:213-228.

20 Lee KW, Kim Y, Kim AM, Helmin K, Nairn AC, Greengard P (2006) Cocaine-induced dendritic

21 spine formation in D1 and D2 dopamine receptor-containing medium spiny neurons in

22 nucleus accumbens. Proc Natl Acad Sci U S A 103:3399-3404.

41

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Levy RJ, Kvajo M, Li Y, Tsvetkov E, Dong W, Yoshikawa Y, Kataoka T, Bolshakov VY,

2 Karayiorgou M, Gogos JA (2015) Deletion of Rapgef6, a candidate schizophrenia

3 susceptibility gene, disrupts amygdala function in mice. Transl Psychiatry 5:e577.

4 Lobo MK, Nestler EJ (2011) The striatal balancing act in drug addiction: distinct roles of direct

5 and indirect pathway medium spiny neurons. Frontiers in neuroanatomy 5:41.

6 Lobo MK, Covington HE, 3rd, Chaudhury D, Friedman AK, Sun H, Damez-Werno D, Dietz DM,

7 Zaman S, Koo JW, Kennedy PJ, Mouzon E, Mogri M, Neve RL, Deisseroth K, Han MH,

8 Nestler EJ (2010) Cell type-specific loss of BDNF signaling mimics optogenetic control of

9 cocaine reward. Science 330:385-390.

10 Lu L, Koya E, Zhai H, Hope BT, Shaham Y (2006) Role of ERK in cocaine addiction. Trends

11 Neurosci 29:695-703.

12 Maeta K, Hattori S, Ikutomo J, Edamatsu H, Bilasy SE, Miyakawa T, Kataoka T (2018)

13 Comprehensive behavioral analysis of mice deficient in Rapgef2 and Rapgef6, a subfamily

14 of guanine nucleotide exchange factors for Rap small possessing the Ras/Rap-

15 associating domain. Mol Brain 11:27.

16 Mazzucchelli C, Vantaggiato C, Ciamei A, Fasano S, Pakhotin P, Krezel W, Welzl H, Wolfer DP,

17 Pages G, Valverde O, Marowsky A, Porrazzo A, Orban PC, Maldonado R, Ehrengruber

18 MU, Cestari V, Lipp HP, Chapman PF, Pouyssegur J, Brambilla R (2002) Knockout of

19 ERK1 MAP kinase enhances synaptic plasticity in the striatum and facilitates striatal-

20 mediated learning and memory. Neuron 34:807-820.

21 Miller CA, Marshall JF (2005) Molecular substrates for retrieval and reconsolidation of cocaine-

22 associated contextual memory. Neuron 47:873-884.

42

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Nagai T, Yoshimoto J, Kannon T, Kuroda K, Kaibuchi K (2016a) Phosphorylation Signals in

2 Striatal Medium Spiny Neurons. Trends Pharmacol Sci 37:858-871.

3 Nagai T, Nakamuta S, Kuroda K, Nakauchi S, Nishioka T, Takano T, Zhang X, Tsuboi D,

4 Funahashi Y, Nakano T, Yoshimoto J, Kobayashi K, Uchigashima M, Watanabe M, Miura

5 M, Nishi A, Kobayashi K, Yamada K, Amano M, Kaibuchi K (2016b) Phosphoproteomics

6 of the Dopamine Pathway Enables Discovery of Rap1 Activation as a Reward Signal In

7 Vivo. Neuron 89:550-565.

8 Nestler EJ (2016) Reflections on: "A general role for adaptations in G- and the cyclic AMP

9 system in mediating the chronic actions of morphine and cocaine on neuronal function".

10 Brain Res 1645:71-74.

11 Nestler EJ, Luscher C (2019) The Molecular Basis of Drug Addiction: Linking Epigenetic to

12 Synaptic and Circuit Mechanisms. Neuron 102:48-59.

13 Park JM, Hu JH, Milshteyn A, Zhang PW, Moore CG, Park S, Datko MC, Domingo RD, Reyes

14 CM, Wang XJ, Etzkorn FA, Xiao B, Szumlinski KK, Kern D, Linden DJ, Worley PF

15 (2013) A prolyl-isomerase mediates dopamine-dependent plasticity and cocaine motor

16 sensitization. Cell 154:637-650.

17 Pascoli V, Cahill E, Bellivier F, Caboche J, Vanhoutte P (2014) Extracellular signal-regulated

18 protein kinases 1 and 2 activation by addictive drugs: a signal toward pathological

19 adaptation. Biol Psychiatry 76:917-926.

20 Pascoli V, Besnard A, Herve D, Pages C, Heck N, Girault JA, Caboche J, Vanhoutte P (2011)

21 Cyclic adenosine monophosphate-independent tyrosine phosphorylation of NR2B

22 mediates cocaine-induced extracellular signal-regulated kinase activation. Biol Psychiatry

23 69:218-227.

43

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Ravni A, Vaudry D, Gerdin MJ, Eiden MV, Falluel-Morel A, Gonzalez B, Vaudry H, Eiden LE

2 (2008) A cAMP-dependent, PKA-independent signaling pathway mediating

3 neuritogenesis through Egr1 in PC12 cells. Mol Pharmacol 73:1688-1708.

4 Rebec GV, Sun W (2005) Neuronal substrates of relapse to cocaine-seeking behavior: role of

5 prefrontal cortex. J Exp Anal Behav 84:653-666.

6 Ren Z, Sun WL, Jiao H, Zhang D, Kong H, Wang X, Xu M (2010) Dopamine D1 and N-methyl-

7 D-aspartate receptors and extracellular signal-regulated kinase mediate neuronal

8 morphological changes induced by repeated cocaine administration. Neuroscience 168:48-

9 60.

10 Robinson TE, Berridge KC (2000) The psychology and neurobiology of addiction: an incentive-

11 sensitization view. Addiction 95 Suppl 2:S91-117.

12 Russo SJ, Nestler EJ (2013) The brain reward circuitry in mood disorders. Nat Rev Neurosci

13 14:609-625.

14 Santini E, Feyder M, Gangarossa G, Bateup HS, Greengard P, Fisone G (2012) Dopamine- and

15 cAMP-regulated phosphoprotein of 32-kDa (DARPP-32)-dependent activation of

16 extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin complex

17 1 (mTORC1) signaling in experimental parkinsonism. J Biol Chem 287:27806-27812.

18 Santini E, Valjent E, Usiello A, Carta M, Borgkvist A, Girault JA, Herve D, Greengard P, Fisone

19 G (2007) Critical involvement of cAMP/DARPP-32 and extracellular signal-regulated

20 protein kinase signaling in L-DOPA-induced dyskinesia. J Neurosci 27:6995-7005.

21 Shaywitz AJ, Greenberg ME (1999) CREB: A stimulus-induced transcription factor activated by

22 a diverse array of extracellular signals. Annu Rev Biochem 68:821-861.

44

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Shuen JA, Chen M, Gloss B, Calakos N (2008) Drd1a-tdTomato BAC transgenic mice for

2 simultaneous visualization of medium spiny neurons in the direct and indirect pathways of

3 the basal ganglia. J Neurosci 28:2681-2685.

4 Soares-Cunha C, de Vasconcelos NAP, Coimbra B, Domingues AV, Silva JM, Loureiro-Campos

5 E, Gaspar R, Sotiropoulos I, Sousa N, Rodrigues AJ (2019) Nucleus accumbens medium

6 spiny neurons subtypes signal both reward and aversion. Mol Psychiatry.

7 Sun WL, Zhou L, Hazim R, Quinones-Jenab V, Jenab S (2007) Effects of acute cocaine on ERK

8 and DARPP-32 phosphorylation pathways in the caudate-putamen of Fischer rats. Brain

9 Res 1178:12-19.

10 Svenningsson P, Nishi A, Fisone G, Girault JA, Nairn AC, Greengard P (2004) DARPP-32: an

11 integrator of neurotransmission. Annu Rev Pharmacol Toxicol 44:269-296.

12 Tejeda HA, Wu J, Kornspun AR, Pignatelli M, Kashtelyan V, Krashes MJ, Lowell BB, Carlezon

13 WA, Jr., Bonci A (2017) Pathway- and Cell-Specific Kappa-Opioid Receptor Modulation

14 of Excitation-Inhibition Balance Differentially Gates D1 and D2 Accumbens Neuron

15 Activity. Neuron 93:147-163.

16 Uhl GR, Koob GF, Cable J (2019) The neurobiology of addiction. Ann N Y Acad Sci.

17 Valjent E, Corvol JC, Trzaskos JM, Girault JA, Herve D (2006a) Role of the ERK pathway in

18 psychostimulant-induced locomotor sensitization. BMC Neurosci 7:20.

19 Valjent E, Corbille AG, Bertran-Gonzalez J, Herve D, Girault JA (2006b) Inhibition of ERK

20 pathway or protein synthesis during reexposure to drugs of abuse erases previously learned

21 place preference. Proc Natl Acad Sci U S A 103:2932-2937.

45

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

1 Valjent E, Corvol JC, Pages C, Besson MJ, Maldonado R, Caboche J (2000) Involvement of the

2 extracellular signal-regulated kinase cascade for cocaine-rewarding properties. J Neurosci

3 20:8701-8709.

4 Valjent E, Bertran-Gonzalez J, Aubier B, Greengard P, Herve D, Girault JA (2010) Mechanisms

5 of locomotor sensitization to drugs of abuse in a two-injection protocol.

6 Neuropsychopharmacology 35:401-415.

7 Valjent E, Aubier B, Corbille AG, Brami-Cherrier K, Caboche J, Topilko P, Girault JA, Herve D

8 (2006c) Plasticity-associated gene Krox24/Zif268 is required for long-lasting behavioral

9 effects of cocaine. J Neurosci 26:4956-4960.

10 Valjent E, Pascoli V, Svenningsson P, Paul S, Enslen H, Corvol JC, Stipanovich A, Caboche J,

11 Lombroso PJ, Nairn AC, Greengard P, Herve D, Girault JA (2005) Regulation of a protein

12 phosphatase cascade allows convergent dopamine and glutamate signals to activate ERK

13 in the striatum. Proc Natl Acad Sci USA 102:491-496.

14 Xu M, Hu XT, Cooper DC, Moratalla R, Graybiel AM, White FJ, Tonegawa S (1994) Elimination

15 of cocaine-induced hyperactivity and dopamine-mediated neurophysiological effects in

16 dopamine D1 receptor mutant mice. Cell 79:945-955.

17 Zhang Y, Kurup P, Xu J, Carty N, Fernandez SM, Nygaard HB, Pittenger C, Greengard P,

18 Strittmatter SM, Nairn AC, Lombroso PJ (2010) Genetic reduction of striatal-enriched

19 tyrosine phosphatase (STEP) reverses cognitive and cellular deficits in an Alzheimer's

20 disease mouse model. Proc Natl Acad Sci U S A 107:19014-19019.

21

22

23

46 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 1 A NS-1 (Rapgef2 +/+) NS-1 (Rapgef2 -/-) 8-CPT-cAMP: -- + + -- + + Egr-1

phospho-ERK

pan-ERK

B phospho-ERK Egr-1 ** 6 * ** 4 ** ns 2 ns

Relative protein level 0 8-CPT-cAMP: (-) (+) (-) (+) (-) (+) (-) (+) NS-1 NS-1 NS-1 NS-1 (Rapgef2 +/+) (Rapgef2 -/-) (Rapgef2 +/+) (Rapgef2 -/-) bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 2

A Rapgef2/Bassoon/D API mPFC a a1 B Rapgef2/PSD95/DAPI DAPI PSD95 Rapgef2 Rapgef2/PSD95/DAPI a mPFC

a1 a2

a2

mPFC NAc b b1

b2 b2 NAc b1 b

BLA c c1

BLA c1

c2

c c2 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 3 A B pERK/DAPI pERK D1R-tdTomato/pERK Vehicle a 1 aca a 3 2

Vehicle+SKF81297 Vehicle+SKF81297 b 1

aca b

U0126+SKF81297 c 2

aca c

EL1101+SKF81297 d 3

aca d Figure 4 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. A flox cKO B ACC ACC AAV9-hSyn-eGFP (ctrl virus) or mPFC mPFC AAV9-hSyn-cre-eGFP (cre virus)

PL PL II/III V VI II/III V VI Rapgef2

IL IL

4 weeks NAc DAPI BLA BLA Rapgef2 flox or C57BL/6J WT mice Rapgef2/ C flox/ctrl virus flox/cre virus C57BL/6J WT/ cre virus NAc NAc Rapgef2 Rapgef2/GFP/DA PI Rapgef2 Rapgef2/GFP/DA PI Rapgef2 Rapgef2/GFP/DA PI DAPI Rapgef2/

NAc NAc DAPI

1.5 Rapgef2/ 1.2

1.2 0.9

0.6 0.8 0.3 * 0.4 * * Relative Rapgef2 IR 0 (ctrl virus) (cre virus) (cre virus)

Relative Rapgef2 IR 0 flox cKO flox cKO flox cKO flox flox WT mPFC NAc BLA Figure 5

A D1R-tdTomato B flox cKO SAL COC pERK/TdTomato pERK/TdTomato pERK/TdTomato SAL COC SAL COC ACC a b c ACC mPFC

TdTomato PL / mPFC mPFC DAPI II/III V VI a NAc BLA PL

pERK IL II III V VI

II/III V VI II/III V VI II/III V VI II/III V VI pERK/ IL aca aca

NAc TdTomato / aca II III V VI DAPI

pERK b

NAc

pERK/ TdTomato bioRxiv / preprintBLA doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder.c ThisII article III is a US V Government VI work. It is not subject to copyright under 17 USC 105

and is also made available for use under a CC0 license. pERK

200um BLA pERK C Egr-1/zif268 IR flox cKO SAL COC SAL COC D phospho-ERK Egr-1/zif268 mPFC mPFC

4 ERK

- * 3 * ACC

3 IR 1 PL - IL 2 mPFC 2 1 V III/II 1 V III/II V III/II V III/II

0 EgrRelative 0 positive cell number number positive cell Bregma 1.70 mm phospho Relative flox cKO flox cKO flox cKO flox cKO SAL COC SAL COC core core core core

NAc 25 NAc 5 NAc ERK

shell shell shell shell - 1 IR IR 1 4 20 - 15 3 10 2 NAc 5 1

0 Relative Egr 0 positive cell number positive number cell Bregma 1.42mm phospho Relative flox cKO flox cKO flox cKO flox cKO SAL COC SAL COC BLA

6 BLA 3 BLA

ERK * *

-

1 IR IR 1 - 4 2 NAc_SAL NAc_COC 2 1 E BLA

R R EgrRelative L L number positive cell 0 0 aca aca Bregma -2.18 mm phospho Relative flox cKO flox cKO flox cKO flox cKO aca SAL COC SAL COC aca ctrl

pERK virus F ctrl virus cre virus S S C C S S C C L R L R aca aca Egr-1 aca aca cre virus GAPDH pERK

Egr-1 pERK * 2.5 * 12 * 2 1 IR IR 1 +

* - ns 9 1.5

pERK 6 1 ns

neuron # neuron 3 0.5 RelativeEgr

Relative 0 0 ctrl virus cre virus ctrl virus cre virus ctrl virus cre virus ctrl virus cre virus SAL COC SAL COC bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 6

A Locomotor Sensitization experimental design C SAL Avg COC1 COC2 (Two-Injection Protocol) ** ** * 25000 ** AAV9-hSyn-eGFP or * ns ** AAV9-hSyn-cre-eGFP Day 1~4 * NAc infusion SAL/daily 20000

15000 Day 5 Day 6 COC1 3~4 weeks COC2 10000

B break beam # of SAL Avg 5000 2000 flox_ctrl virus in NAc flox_cre virus in NAc 1600 Saline, ip flox 0 flox_ctrl virus in NAC flox_cre virus in NAC flox cKO 1200 cKO

800

400 ctrl virus cre virus # of # of break beam D 0 5 15 25 35 45 55 65 75 85 (min)

2000 COC 1

1600 Cocaine, ip Bregma 0.86 mm (15 mg/kg) 1200

800

# of # of break beam Bregma 1.10 mm 400

0 5 15 25 35 45 55 65 75 85 (min) Bregma 1.34 mm COC 2 2000 Cocaine, ip 1600 (15 mg/kg)

1200 Bregma 1.54 mm

800

# of # of break beam 400

0 Bregma 1.78 mm 5 15 25 35 45 55 65 75 85 (min) bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 7

A Conditioned Place Preference (CPP) experimental design Day 2 Day 11 D Ctrl virus Cre virus PreconditioningTest postconditioning Test

AAV9-hSyn-eGFP or AAV9-hSyn-cre-eGFP Day 1 Day 3~10 NAc infusion exploration SAL/COC (10mg/kg) conditioning

Bregma 0.86 mm

S1 C1 S2 C2 S3 C3 S4 C4 (COC group) 3~4 weeks S1 S1’ S2 S2’ S3 S3’ S4 S4’ (SAL group) ns Bregma 1.10 mm B C * 800 ctrl virus SAL * * 800 flox SAL ** ctrl virus COC ns flox COC * ** 600 cre virus SAL 600 cKO SAL Bregma 1.34 mm cre virus COC cKO COC 400 400

200 ns 200 ns 0 0 Bregma 1.54 mm

-200 -200 CPP scores (sec) -400 CPP scores (sec) -400 Bregma 1.78 mm -600 -600 ns ns -800 -800 preconditioning postconditioning preconditioning postconditioning bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 8

SAL COC A pERK pERK/GFP pERK pERK/GFP

ctrl virus

Phospho- ERK cre virus

pCREB pCREB/GFP pCREB pCREB/GFP

ctrl virus

Phospho- CREB

cre virus

B pERK pCREB * ** 20 * 4 ns 16 signal IR 3 + neuron # # neuron + 12 2

pCREB ns pERK 8 ns that arethat GFP+ 4 GFP+neurons in 1 Relative Relative 0 0 ctrl virus cre virus ctrl virus cre virus ctrl virus cre virus ctrl virus cre virus SAL COC SAL COC Figure 9

New model for ERK activation in D1 dopaminoceptive neurons

D1R NMDAR AC NCS- GluN1 GluN2B Gs/olf Rapgef2 Ca2+

cAMP PKA Rap1 Ras-GRF1

Ras-GRP2

DARPP32 PP1 MEK

STEP STEP ERK

cocaine-induced dopamine increase

bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which basal glutamate level was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. phosphorylation activation inhibition

Elk-1 ERK

CREB ERK gene regulation H3H3H3

Elk-1 Egr-1 SRF SRF chromatin CRE SRE remodeling

gene regulation H3H3H3 CREB

chromatin CRE remodeling

other long-term effects plasticity for behaviors bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 4-1

A +/- B Drd1-cre :: Rosa-eGFP:: Drd1-cre+/-::Rapgef2cko/cko +/- Drd1-cre :: Rosa- Rapgef2cko/cko with D1 agonist SKF 81297 eGFP NAc aca aca shell aca core core shell core shell

pERK

NAc/shell NAc/shell GFP/Rapgef2 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 5-1

flox cKO SAL SKF81297 SAL SKF81297 ACC

mPFC D API PL

pERK/ II/III V VI II/III V VI II/III V VI II/III V VI IL

aca D API NAc pERK/ D API BLA pERK/ bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 5-2 Drd1-cre+/-:: Rosa-eGFP Drd2-cre+/-::Rosa-eGFP A Rapgef2/GFP/DAPI Rapgef2/GFP/DAPI NAc NAc

B saline eticlopride pERK pERK/GFP pERK pERK/GFP pERK NAc NAc pERK+ pERK+ & GFP+ ctrl * virus ns 12

9 + neuron neuron # + 6 NAc NAc ns

pERK 3

cre 0 virus ctrl virus cre virus ctrl virus cre virus Relative Relative SAL SAL eti eti bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 6-1

A

10000 20 flox cKO 35 8000 30 15 25 6000 20 10 4000 15 Arms entries Arms 10 2000 5 5 Time in open arms (%) 0 Total Total distance traveled (mm) 0 0 flox cKO Entries in Entries in flox cKO Open Arms Closed Arms

B ctrl virus cre virus 8000 14 20 12 6000 10 15

4000 8 10 6

2000 entries Arms 4 5

2 Time in open arms (%) Total Total distance traveled (mm) 0 0 0 ctrl virus cre virus Entries in Open Entries in ctrl virus cre virus Arms Closed Arms bioRxiv preprint doi: https://doi.org/10.1101/2020.07.02.184754; this version posted July 4, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license.

Figure 6-2 C57BL/6N with cre viral injection in NAc

A SAL_Avg COC1 1500 COC2 SAL/COC 1200

900

600

300

# of breaks beam 0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 (min)

B C57BL/6J with cre viral injection in NAc 2500 SAL_Avg COC1 2000 COC2 SAL/COC 1500

1000

# of breaks beam 500

0 5 10 15 20 25 30 35 40 45 50 55 60 65 70 75 80 85 90 C (min) 25000 * * SAL Avg * * COC1 20000 COC2

15000

10000

# of beam breaks beam # of 5000

0 C57BL/6N_cre virus in NAc C57BL/6J_cre virus in NAc