49
Accepted manuscripts are peer-reviewed but have not been through the copyediting, formatting, or proofreading process. Copyright © 2019 the authors This Accepted Manuscript has not been copyedited and formatted. The final version may differ from this version. A link to any extended data will be provided when the final version is posted online. Research Articles: Cellular/Molecular Activation of dopamine receptor 2 (DRD2) prompts transcriptomic and metabolic plasticity in glioblastoma Seamus P. Caragher, Jack M. Shireman, Mei Huang, Jason Miska, Fatemeh Atashi, Shivani Baisiwala, Cheol Hong Park, Miranda R. Saathoff, Louisa Warnke, Ting Xiao, Maciej S. Lesniak, C. David James, Herbert Meltzer 1 , Andrew K. Tryba 2 and Atique U Ahmed 1 Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL USA 60616 1 Department of Psychiatry, Feinberg School of Medicine, Northwestern University, Chicago, IL USA 60616. 2 Department of Pediatrics, Pritzker School of Medicine, University of Chicago, Chicago, IL USA 60637 https://doi.org/10.1523/JNEUROSCI.1589-18.2018 Received: 24 June 2018 Revised: 17 December 2018 Accepted: 28 December 2018 Published: 16 January 2019 Author contributions: S.P.C., A.K.T., and A.A. designed research; S.P.C., J.M.S., M.H., J.M., F.A., S.B., C.H.P., M.S., L.W., and A.K.T. performed research; S.P.C., J.M.S., M.H., J.M., S.B., and C.H.P. contributed unpublished reagents/analytic tools; S.P.C., J.M.S., J.M., C.H.P., T.X., C.D.J., A.K.T., and A.A. analyzed data; S.P.C. wrote the first draft of the paper; J.M.S., M.S.L., C.D.J., H.Y.M., A.K.T., and A.A. edited the paper; A.A. wrote the paper. Conflict of Interest: The authors declare no competing financial interests. This work was supported by the National Institute of Neurological Disorders and Stroke grant 1R01NS096376, the American Cancer Society grant RSG-16-034-01-DDC (to A.U.A.) and National Cancer Institute grant R35CA197725 (to M.S.L) and P50CA221747 SPORE for Translational Approaches to Brain Cancer. We would like to thank Meijing Wu for the statistical analysis for the preparation of this manuscript. Correspondence should be addressed to To whom correspondence should be addressed: Atique U Ahmed, Department of Neurological Surgery, Room 3-112, 303 E Superior Street, Chicago, IL 60616, Email: [email protected] Phone: +1 (312) 503-3552 Cite as: J. Neurosci 2019; 10.1523/JNEUROSCI.1589-18.2018 Alerts: Sign up at www.jneurosci.org/alerts to receive customized email alerts when the fully formatted version of this article is published.

Activation of dopamine receptor 2 (DRD2) prompts

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Activation of dopamine receptor 2 (DRD2) prompts

Accepted manuscripts are peer-reviewed but have not been through the copyediting, formatting, or proofreadingprocess.

Copyright © 2019 the authors

This Accepted Manuscript has not been copyedited and formatted. The final version may differ fromthis version. A link to any extended data will be provided when the final version is posted online.

Research Articles: Cellular/Molecular

Activation of dopamine receptor 2 (DRD2) prompts transcriptomic andmetabolic plasticity in glioblastomaSeamus P. Caragher, Jack M. Shireman, Mei Huang, Jason Miska, Fatemeh Atashi, Shivani Baisiwala,Cheol Hong Park, Miranda R. Saathoff, Louisa Warnke, Ting Xiao, Maciej S. Lesniak, C. David James,

Herbert Meltzer1, Andrew K. Tryba2 and Atique U Ahmed

1Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL USA606161Department of Psychiatry, Feinberg School of Medicine, Northwestern University, Chicago, IL USA 60616.2Department of Pediatrics, Pritzker School of Medicine, University of Chicago, Chicago, IL USA 60637

https://doi.org/10.1523/JNEUROSCI.1589-18.2018

Received: 24 June 2018

Revised: 17 December 2018

Accepted: 28 December 2018

Published: 16 January 2019

Author contributions: S.P.C., A.K.T., and A.A. designed research; S.P.C., J.M.S., M.H., J.M., F.A., S.B.,C.H.P., M.S., L.W., and A.K.T. performed research; S.P.C., J.M.S., M.H., J.M., S.B., and C.H.P. contributedunpublished reagents/analytic tools; S.P.C., J.M.S., J.M., C.H.P., T.X., C.D.J., A.K.T., and A.A. analyzed data;S.P.C. wrote the first draft of the paper; J.M.S., M.S.L., C.D.J., H.Y.M., A.K.T., and A.A. edited the paper; A.A.wrote the paper.

Conflict of Interest: The authors declare no competing financial interests.

This work was supported by the National Institute of Neurological Disorders and Stroke grant 1R01NS096376,the American Cancer Society grant RSG-16-034-01-DDC (to A.U.A.) and National Cancer Institute grantR35CA197725 (to M.S.L) and P50CA221747 SPORE for Translational Approaches to Brain Cancer. We wouldlike to thank Meijing Wu for the statistical analysis for the preparation of this manuscript.

Correspondence should be addressed to To whom correspondence should be addressed: Atique UAhmed, Department of Neurological Surgery, Room 3-112, 303 E Superior Street, Chicago, IL 60616, Email:[email protected] Phone: +1 (312) 503-3552

Cite as: J. Neurosci 2019; 10.1523/JNEUROSCI.1589-18.2018

Alerts: Sign up at www.jneurosci.org/alerts to receive customized email alerts when the fully formatted versionof this article is published.

Page 2: Activation of dopamine receptor 2 (DRD2) prompts

1

Activation of dopamine receptor 2 (DRD2) prompts transcriptomic and metabolic 1

plasticity in glioblastoma 2

Abbreviated title: DRD2 activates stemness and alters metabolism in GBM 3

Seamus P. Caragher*, Jack M. Shireman*, Mei Huang, Jason Miska, Fatemeh Atashi, Shivani 4

Baisiwala, Cheol Hong Park, Miranda R. Saathoff, Louisa Warnke, Ting Xiao, Maciej S. 5

Lesniak, C. David James, Herbert Meltzer1, Andrew K. Tryba2, Atique U Ahmed 6

7

Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 8

Chicago, IL USA 60616, 1Department of Psychiatry, Feinberg School of Medicine, 9

Northwestern University, Chicago, IL USA 60616. 2Department of Pediatrics, Pritzker School of 10

Medicine, University of Chicago, Chicago, IL USA 60637 11

*These Authors Contributed Equally to this Manuscript 12 13 To whom correspondence should be addressed: 14 Atique U Ahmed 15 Department of Neurological Surgery 16 Room 3-112 17 303 E Superior Street 18 Chicago, IL 60616 19 Email: [email protected] 20 Phone: +1 (312) 503-3552 21 22 Pages: 36 23 Figures and tables: 6 24

Word counts: Abstract: 163 Introduction: 515 Discussion: 1921 25

Conflicts of Interest: The authors declare no competing interests. 26

Acknowledgement: This work was supported by the National Institute of Neurological 27 Disorders and Stroke grant 1R01NS096376, the American Cancer Society grant RSG-16-034-28 01-DDC (to A.U.A.) and National Cancer Institute grant R35CA197725 (to M.S.L) and 29 P50CA221747 SPORE for Translational Approaches to Brain Cancer. We would like to thank 30 Meijing Wu for the statistical analysis for the preparation of this manuscript. 31 32

Page 3: Activation of dopamine receptor 2 (DRD2) prompts

2

Abstract 33

Glioblastoma (GBM) is one of the most aggressive and lethal tumor types. Evidence 34

continues to accrue indicating that the complex relationship between GBM and the brain 35

microenvironment contributes to this malignant phenotype. However, the interaction between 36

GBM and neurotransmitters, signaling molecules involved in neuronal communication, remains 37

incompletely understood. Here we examined, using human patient derived xenograft lines, how 38

the monoamine dopamine influences GBM cells. We demonstrate that GBM cells express 39

DRD2, with elevated expression in the glioma-initiating cell (GIC) population. Stimulation of 40

DRD2 caused neuron-like depolarization exclusively in GICs. In addition, long-term activation 41

of DRD2 heightened the sphere-forming capacity of GBM cells, as well as tumor engraftment 42

efficiency in both male and female mice. Mechanistic investigation revealed that DRD2 43

signaling activates the hypoxia response and functionally alters metabolism. Finally, we found 44

that GBM cells synthesize and secrete dopamine themselves, suggesting a potential autocrine 45

mechanism. These results identify dopamine signaling as a potential therapeutic target in GBM 46

and further highlight neurotransmitters as a key feature of the pro-tumor microenvironment. 47

Significance Statement: 48

This work offers critical insight into the role of the neurotransmitter dopamine in the progression 49

of GBM. We show that dopamine induces specific changes in the state of tumor cells, 50

augmenting their growth and shifting them to a more stem-cell like state. Further, our data 51

illustrate that dopamine can alter the metabolic behavior of GBM cells, increasing glycolysis. 52

Finally, this work demonstrates that GBM cells, including tumor samples from patients, can 53

synthesize and secrete dopamine, suggesting an autocrine signaling process underlying these 54

Page 4: Activation of dopamine receptor 2 (DRD2) prompts

3

results. These results describe a novel connection between neurotransmitters and brain cancer, 55

further highlighting the critical influence of the brain milieu on GBM. 56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

Page 5: Activation of dopamine receptor 2 (DRD2) prompts

4

Introduction 78

Glioblastoma (GBM) is the most common and aggressive primary malignant brain tumor 79

afflicting adults, with standard of care treatment leading to a median survival of only 15 months 80

(Alifieris and Trafalis 2015). Recent clinical trials have shown increased survival to 81

approximately 21 months (Stupp et al. 2017). A key factor limiting the efficacy of treatment is 82

the remarkable plasticity exhibited by GBM cells, which allows them to effectively adapt to 83

changes in the microenvironment induced by conventional radiation and chemotherapy (Safa et 84

al. 2015; Olmez et al. 2015). Our group and others have shown that cellular plasticity processes 85

enable GBM cells to adopt many phenotypes, including the glioma initiating cell (GIC) state, 86

characterized by a neural stem cell-like expression profile and heightened resistance to therapy 87

(Safa et al. 2015; Olmez et al. 2015; Dahan et al. 2014; Lee et al. 2016). Given the dynamic 88

nature of this process, a key question is what mechanisms regulate and control cellular plasticity. 89

Several factors have been shown to activate GBM plasticity, including hypoxia, acidity, and 90

metabolic changes (Hardee et al. 2012; Heddleston et al. 2009; Hjelmeland et al. 2011; Soeda et 91

al. 2009; Tamura et al. 2013). All of these factors are influenced by the dynamic 92

microenvironmental milieu occupied by GBM cells. Therefore, further analysis of how 93

environmental factors specific to the central nervous system (CNS) affect tumors are critical for 94

devising novel therapies for patients with GBM. 95

96

The GBM microenvironment is unique for a multitude of reasons, including the range of CNS-97

specific factors, such as neurotransmitters and neurotrophins, known to influence cell 98

proliferation and differentiation. Given the evidence that plasticity can be induced in GBM tumor 99

cells by a variety of microenvironmental factors, including oxygen availability and acidity 100

Page 6: Activation of dopamine receptor 2 (DRD2) prompts

5

(Hjelmeland et al. 2011; Soeda et al. 2009; Evans et al. 2004), it is logical that CNS-specific cues 101

like neurotransmitters may also influence GBM cellular behavior. Indeed, evidence is 102

accumulating that CNS-specific cues contribute to the onset and progression of GBM (Charles et 103

al. 2011). It has been shown, for example, that factors secreted from neurons such as neuroligin-104

3 directly augment GBM cell growth (Venkatesh et al. 2015; Venkatesh et al. 2017). Further, 105

Villa and colleagues have demonstrated that GBM cells utilize cholesterol secreted by healthy 106

astrocytes to enhance their growth (Villa et al. 2016). In addition, glutamate, a neurotransmitter, 107

has been shown to augment GBM growth via effects on EGFR signaling (Schunemann et al. 108

2010). 109

110

One neurotransmitter family likely to contribute to the pro-tumor brain microenvironment in 111

GBM is the monoamines (dopamine, serotonin, and norepinephrine), which are critical to 112

behavior, emotion, and cognition (Beaulieu and Gainetdinov 2011; Lammel, Lim, and Malenka 113

2014). A recent review highlighted the potential influence of monoamines on GBM tumors 114

(Caragher et al. 2017). Critically, multiple studies have shown that monoamine signaling 115

influences the behavior and phenotype of healthy neural stem cells (NSCs), which have similar 116

expression profiles and functional attributes to GICs (Singh et al. 2004; Galli et al. 2004). In 117

light of these reports, we set out to investigate how DRD2 activation influences GBM signaling 118

and phenotype. 119

120

121

122

123

Page 7: Activation of dopamine receptor 2 (DRD2) prompts

6

124

125

126

Materials and Methods 127

Cell Culture 128

U251 human glioma cell lines were procured from the American Type Culture Collection 129

(Manassas, VA, USA). These cells were cultured in Dulbecco’s Modified Eagle’s Medium 130

(DMEM; HyClone, Thermo Fisher Scientific, San Jose, CA, USA) supplemented with 10% fetal 131

bovine serum (FBS; Atlanta Biologicals, Lawrenceville, GA, USA) and 2% penicillin-132

streptomycin antibiotic mixture (Cellgro, Herdon, VA, USA; Mediatech, Herdon, VA, USA). 133

Patient-derived xenograft (PDX) glioma specimens (GBM43, GBM12, GBM6, GBM5, and 134

GBM39) were obtained from Dr. C David James at Northwestern University and maintained 135

according to published protocols(Hodgson et al. 2009). Notably, these cells are known to 136

represent different subtypes of GBM. GBM43 and GBM12 are proneural, GBM6 and GBM39 137

are classical, and GBM5 is mesenchymal, according to the subtypes established by Verhaak 138

(Verhaak et al. 2010). 139

140

Determination of Dopamine Levels 141

Cell culture supernatant was collected, filtered, and flash frozen. The details of the mass 142

spectrometric/UPLC procedure are described elsewhere (Huang et al. 2014). The LC system 143

(Waters Acquity UPLC, Waters Co., Milford, MA, USA) and a Waters Acquity UPLC HSS T3 144

1.8 μm column were used for separation. The UPLC system was coupled to a triple-quadrupole 145

mass spectrometer (Thermo TSQ Quantum Ultra, Waltham, MA, USA), using ESI in positive 146

Page 8: Activation of dopamine receptor 2 (DRD2) prompts

7

mode. All data were processed by Waters MassLynx 4.0 and Thermo Xcaliber software. Data 147

was acquired and analyzed using Thermo LCquan 2.5 software (Thermo Fisher Scientific, Inc., 148

Waltham, MA, USA). 149

Animals 150

Athymic nude mice (nu/nu; Charles River, Skokie, IL, USA) were housed according to all 151

Institutional Animal Care and Use Committee (IACUC) guidelines and in compliance with all 152

applicable federal and state statutes governing the use of animals for biomedical research. 153

Briefly, animals were housed in shoebox cages with no more than 5 mice per cage in a 154

temperature and humidity-controlled room. Food and water were available ad libitum. A strict 155

12-hour light-dark cycle was maintained. 156

157

MicroArray 158

RNA was extracted from samples using RNeasyI kit according to manufacturer’s 159

instructions (Qiagen). 1000ng of RNA were utilized for MicroArray analysis according to 160

manufacturer’s directions (Ilumina). HumanHT12 (48,000 probes, RefSeq plus EST) was 161

utilized for all microarrays. All microarrays were performed in triplicate. 162

163

Glycolytic measurement 164

GBM 39, 12 and 5 were adhered at a concentration of 0.5-1.5x105 cells per well to XFe 165

96 well culture plates (Agilent; Santa Clara, CA) using CellTak tissue adhesive (Corning; 166

Corning, NY). We then performed a Mito-Stress test as described in the manufacturer’s protocol 167

utilizing a Seahorse Xfe96 well extracellular flux analyzer. Briefly, cells were attached and 168

suspended in XF Base medium supplemented with 2mM L-glutamine (Fisher; Hampton, NH). 169

Page 9: Activation of dopamine receptor 2 (DRD2) prompts

8

After baseline ECAR reading, glucose was injected to a final concentration of 10mM to 170

determine glucose stimulated ECAR rate. Next, oligomycin (1μM final concentration) was 171

injected to determine maximal glycolytic capacity. Lastly, a competitive inhibitor of glycolysis 172

(2-DG; 2-deoxyglucose) was injected (50mM) to validate ECAR as a measure of glycolytic flux. 173

Data was analyzed using Agilent’s proprietary Wave software. 174

175

ChIP Sequencing 176

The PDX line GBM 43 was subject to DMSO or TMZ treatment for 4 days, after which 177

cells were washed, collected, and pelleted. Cells were incubated with anti-H3K27ac antibody, 178

after which crosslinking of protein binding occurred. DNA was sheared and crossing linking was 179

reversed. Resultant DNA was sent to ZYMO Research Facility where it underwent next 180

generation sequencing according to manufactures established protocol. 181

ChIPSeq reads underwent FastQC quality analysis after sequencing and no abnormalities 182

were detected. Alignment was done using Bowtie2 software and peak calling was performed 183

using the MACS2 CallPeak function with p value set to .05. All ChIPSeq data was visualized 184

using Integrated Genomics Viewer (IGV). 185

186

Electrophysiology 187

Whole-cell current-clamp recordings of GBM cells were obtained under visual guidance 188

with a Zeiss Axioscop FS (Zeiss; Jena, Germany); fluorescence was used to target GICs. DRD2 189

agonist was delivered by superfusion of targeted cells using a Picospritzer device (Parker 190

Hannifan; Cleveland, OH, USA). Recordings were made with a Multiclamp 700B (Molecular 191

Devices; Sunnyvale, CA, USA) and Digidata data acquisition devices (Molecular devices) and 192

Page 10: Activation of dopamine receptor 2 (DRD2) prompts

9

digitized at 20,000 samples/s. The baseline membrane potential was corrected for the liquid 193

junction potential calculated using pClamp 10 Software (Molecular Devices). Patch-clamp 194

electrodes were manufactured from filamented borosilicate glass tubes (Clark G150F-4, Warner 195

Instruments). Current-clamp electrodes were filled with an intracellular solution containing the 196

following (in mM): 140 K-gluconate, 1 CaCl2 * 6H2O, 10 EGTA, 2 MgCl2 * 6H2O, 4 Na 197

2ATP, and 10 HEPES with a resistance of ~ 3MΩ. Cells were recorded in artificial CSF (ACSF) 198

containing the following (in mM): 118 NaCl, 3 KCl, 1.5 CaCl2, 1 MgCl2 * 6H2O, 25 NaHCO3, 199

1 NaH2PO4, and 30 D-glucose, equilibrated with medical grade carbogen (95% O2 plus 5% 200

CO2, pH= 7.4). Chemicals for ACSF were obtained from Sigma-Aldrich. Cells were submerged 201

under ACSF, saturated with carbogen in recording chambers (flow rate, 12 ml/min). Experiments 202

were performed at 30°C ±0.7°C using a TC-344B Temperature Regulator with an in-line 203

solution heater (Warner Instruments). 204

205

Statistical Analysis and Study Design 206

All statistical analyses were performed using the GraphPad Prism Software v8.0 (GraphPad 207

Software; San Diego, CA, USA). In general, data are presented as mean (with standard 208

deviation) for continuous variables, and number (percentage) for categorical variables. 209

Differences between two groups were assessed using Student’s t-test or Wilcoxon rank sum test 210

as appropriate. For tumorsphere formation assay a score test for heterogeneity between two 211

groups was used and reported as p-value and Chi-Squared for each group. Difference among 212

multiple groups were evaluated using analysis of variance (ANOVA) with post hoc Tukey’s test, 213

or Mann-Whitney U test followed by Bonferroni correction as appropriate. Survival curves were 214

graphed via Kaplan-Meier method, and compared by log-rank test. All tests were two-sided and 215

Page 11: Activation of dopamine receptor 2 (DRD2) prompts

10

P value <0.05 was considered statistically significant. Experiments such as western blots, 216

microarray, FACS, Glycolytic Measurements, microarray were performed in biological 217

triplicate. HPLC was performed as a biological duplicate across separate experimental conditions 218

data were averaged and then student’s T-Tests were performed to determine significance. All in 219

vivo experiments contained at least three animals per group, with males and females equally 220

represented, and each animal was treated as a technical replicate. 221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

Page 12: Activation of dopamine receptor 2 (DRD2) prompts

11

239

240

241

Results 242

Therapeutic stress induces epigenetic modifications and subsequent increased expression of 243

DRD2 244

In order to begin our investigation of monoamines in GBM, we analyzed the epigenetic 245

status of their receptors during therapeutic stress. We performed a genome-wide ChIP-Seq 246

analysis of PDX GBM43 cells for histone 3 lysine 27 (H3K27) acetylation (ac), a marker of open 247

chromatin and active gene transcription, and H3K27 tri-methylation (me3), a maker of closed 248

chromatin and transcriptional repression. Cells were treated with temozolomide (TMZ, 50μM), 249

equimolar vehicle control DMSO, or radiation (2Gy). ChIP-Seq was performed 96 hours after 250

treatment initiation. Examination of all dopamine, serotonin, and noradrenergic receptors showed 251

that the DRD2 promotor alone possessed enrichment of an H3K27ac mark without any change in 252

H3K27me3 (MACS2 Peak Score: 45.0, fold enrichment: 4.023, P-value = 1.28 E-8) (Figure 1A 253

& B, Extended Data Figure 1-1). Further, analysis of the Cancer Genome Atlas (TCGA) revealed 254

that, of the five dopamine receptors, DRD2 mRNA is present at the highest levels (Extended 255

Data Figure 1-1). 256

To determine if this alteration in histone status of the DRD2 promoter leads to increased 257

protein expression, we analyzed protein levels of all DRDs in PDX GBM cells treated with either 258

vehicle control DMSO or TMZ for 2, 4, 6, or 8 days by western blot. We found that DRD2 was 259

specifically increased after treatment (Figure 1C), compared to the other dopamine receptors. We 260

also performed FACS analysis of DRD2 expression in 3 different PDX cells lines over the same 261

Page 13: Activation of dopamine receptor 2 (DRD2) prompts

12

time course. In line with our western blot data, we observed induction of DRD2 protein 262

expression following TMZ treatment (Figure 1D: GBM6—Day 2, adj. p=.0018; Day 4, adj. 263

p=.0355; Day 6, adj. p=.0001; Day 8, adj. p=.0001. GBM39—Day 2, adj. p=.0131; Day 4, adj. 264

p=.0193; Day 6, adj. p=.2297; Day 8, adj. p=.1585. GBM43—Day 2, adj. p=.0087; Day 4, adj. 265

p=.0001; Day 6, adj. p=.0001; Day 8, adj. p=.0492). To confirm DRD2 expression was not 266

caused by culture conditions, we quantified the expression of DRD2 in orthotopic xenografts 267

from the brains of athymic nude (nu/nu) mice, as well as the presence of DRD2 mRNA in human 268

patient samples (Extended Data Figure 1-1 Panel A). Our results corroborate several recent 269

studies. First, GBM tumors in patients express DRD2; these levels are often elevated compared 270

to healthy brain (Li et al. 2014). DRD2 is known to interact with EGFR signaling, a pathway 271

frequently mutated or hyperactive in GBM (Schunemann et al. 2010; Winkler et al. 2004; 272

Giannini et al. 2005; Mukherjee et al. 2009; Snuderl et al. 2011; Szerlip et al. 2012; Verhaak et 273

al. 2010). Other groups have shown that DRD2 inhibition in conjunction with EGFR antagonists 274

reduced GBM proliferation (O'Keeffe et al. 2009; Li et al. 2014). Based on these results, we 275

selected DRD2 as our primary receptor for further exploration. 276

277

CD133+ glioma initiating cells respond to DRD2 activation with electrophysiological changes 278

We next tested if DRD2 expression correlates with certain cellular phenotypes. First, we 279

examined how DRD2 protein expression is affected by alterations in culture conditions known to 280

promote the GIC state. We examined expression of DRD2 in PDX cells growing either in 1% 281

fetal bovine serum (FBS) containing differentiation condition media or as spheres in 282

supplemented neurobasal media. We found that cells growing as spheres in GIC maintenance 283

media had elevated expression not only of GIC markers like CD133 and Sox2, but also 284

Page 14: Activation of dopamine receptor 2 (DRD2) prompts

13

heightened DRD2 (Figure 2A), indicating that cellular state can influence this receptor’s 285

expression. We next analyzed if DRD2 expression correlates with the expression of CD133, a 286

widely used GIC marker. FACS analysis revealed that DRD2 expression is elevated in the 287

CD133+ population (Figure 2B: GBM6 adj. p=.0001, GBM12 adj. p=.0393, GBM39, adj. 288

p=.0199, GBM43 adj p=.00257). 289

Intrigued by this differential expression, we analyzed functional differences in DRD2 signaling 290

in GICs and non-GICs within a heterogeneous cell population. One of DRD2’s main effects is to 291

alter the polarization of brain cells (Beaulieu and Gainetdinov 2011). We therefore utilized 292

electrophysiology to determine the effects of DRD2 activation on GBM cellular polarization. We 293

have previously developed a GIC-reporter line in which expression of red fluorescent protein 294

(RFP) is under the control of a GIC-specific CD133-promoter. This system enables real-time, 295

faithful identification of tumor-initiating GICs in live cells (Lee et al. 2016). We treated our 296

high-fidelity U251 GIC reporter cell line with the highly specific DRD2 agonist (3'-297

Fluorobenzylspiperone maleate, Tocris) at a dose of 30nM, the estimated concentration of 298

dopamine in cerebrospinal fluid (Floresco et al. 2003; Keefe, Zigmond, and Abercrombie 1993). 299

Electrophysiological recordings revealed that RFP+CD133+ cells responded to DRD2 activation 300

to a greater extent than RFP- non-GICs in the same population (Figure 2C p-value = .0010). 301

Notably, this activation produced hyperpolarization’s similar in amplitude and duration to 302

neuronal responses (Beaulieu and Gainetdinov 2011). We also confirmed that only certain PDX 303

cells respond to the agonist in PDX cells (Figure 2D). These results show that DRD2 receptors in 304

GBM cells are functional and produce GIC-specific effects. 305

306

Activation of DRD2 increases GBM cell self-renewal and tumor engraftment capacity 307

Page 15: Activation of dopamine receptor 2 (DRD2) prompts

14

We next sought to understand the relationship between DRD2 expression and tumor cell 308

phenotype during therapy, specifically self-renewal ability and tumor engraftment capacity. To 309

that end, PDX GBM cells were treated with either DMSO or TMZ. Following four days of 310

exposure, cells were sorted based on DRD2 expression and plated for limiting dilution 311

neurosphere assays. We found that DRD2 positive (DRD+) cells exhibited about 4-fold elevation 312

in sphere formation capacity compared to DRD2 negative (DRD-) cells (Figure 3A, DMSO Pre 313

Treatment p value = .00709 TMZ Pre Treatment p value = .00616), suggesting that the DRD2+ 314

population has enhanced self-renewal capacity. Exposure to physiological dose of TMZ also 315

enhanced the self-renewal capacity in both DRD2+ as compared to DRD2- population. Most 316

importantly, post TMZ therapy enhancement of self-renewal resulted in enhanced tumor 317

engraftment capacity in the orthotopic xenograft model as 100% of the mice developed tumor 318

when 500 DRD2+ cells were implanted in the brain of the immunocompromised nu/nu mice 319

(Figure 3B). 320

Based on our data, as well as recently reported data demonstrating that DRD2 signaling 321

influences normal progenitor phenotype and differentiation (Baker, Baker, and Hagg 2004; 322

O'Keeffe et al. 2009; Winner et al. 2009; Ohtani et al. 2003), we next examined if the observed 323

self-renewal capacity of DRD2+ cells was indeed DRD2 dependent and if tumor subtype 324

influences such characteristics. Multiple subtypes of PDX cell lines were plated in the presence 325

of either vehicle control DMSO or 30nM DRD2 agonist and, after 8 days, neurospheres were 326

counted. Our results demonstrate that, for three of the PDX lines tested, DRD2 activation 327

increases sphere-forming capacity (Figure 3C GBM5 p-value = .00239, GBM6 p-value = .00362, 328

GBM39 p-value = .0138 ). GBM12, however, did not respond to the agonist, thus indicating may 329

be some subtype specific variability when responding to DRD2 signaling (Extended Data Figure 330

Page 16: Activation of dopamine receptor 2 (DRD2) prompts

15

3-1) In order to confirm this increase in GIC, we analyzed the expression of several key GIC 331

markers following treatment with the DRD2 agonist and found that cell lines that responded to 332

the agonist by increasing sphere formation also showed increased expression of genes associated 333

with stemness (Extended Data Figure 3-1). Importantly, treatment did not increase cell 334

proliferation as measured by the Ki67 FACS, suggesting our result comes from cellular plasticity 335

and not increased GIC proliferation (Extended Data figure 3-1). In addition, inhibition of DRD2 336

by chlorpromazine (CPM, 1μM), a selective DRD2 antagonist(Beaulieu and Gainetdinov 2011) 337

in agonist-responsive cell lines GBM6 and GBM39, markedly reduced sphere formation, as 338

measured by neurosphere assays (Figure 3D GBM6 p-value= 1.42E-5, GBM 39 p-value= 4.93E-339

10). Finally, viral knockout of DRD2 showed significant reduction in sphere forming capacity in 340

GBM 43 across two different SH-RNA constructs (Figure 3E sh2 p-value = 1.19E-10, sh3 p-341

value = 4.53E-10). 342

343

Glioblastoma cells express tyrosine hydroxylase (TH) and secrete dopamine 344

Our data indicate that DRD2 activation alters the phenotype of GBM cells. We next 345

investigated the possibility that this DRD2 activation occurs via autocrine release of its natural 346

ligand, dopamine. Production of monoamines like dopamine is usually a tightly regulated 347

process limited to specific dopaminergic neuronal populations. Critically, all our experiments 348

were performed in purified monocultures of cancer cells in the absence of dopaminergic neurons, 349

raising the question of the mechanism of DRD2 activation. To investigate the possibility that 350

tumor cells synthesize their own dopamine, we first examined expression of tyrosine 351

hydroxylase (TH), the rate-limiting enzyme of dopamine synthesis, in PDX GBM cells (Figure 352

4A). To confirm this expression occurs in vivo, we intracranially implanted PDX GBM cells into 353

Page 17: Activation of dopamine receptor 2 (DRD2) prompts

16

athymic nude mice; upon visible signs of tumor burden, mice were sacrificed and whole brains 354

were obtained. FACS analysis revealed that human leukocyte antigen (HLA+) tumor cells do in 355

fact express TH in vivo (Figure 4B). Examination of GlioVis data (Bowman et al. 2017) also 356

demonstrated that patient GBM samples express TH and other enzymes necessary for dopamine 357

synthesis, as well as transporters for vesicle loading and reuptake (Extended Data Figure 4-1). 358

To determine if PDX monocultures were generating dopamine, we assayed dopamine levels 359

using high performance liquid chromatography with mass spectrometry (HPLC-MS). 360

Conditioned media of PDX monocultures were found to have dopamine levels of 20-65 nM, 361

between 10 and 30 times greater than cell free media (Figure 4C). The levels of dopamine were 362

relatively stable in samples taken over 8 days, suggesting that the PDX cells produce and 363

maintain basal dopamine synthesis. Next, we quantified dopamine in human tumor samples. A 364

portion of cortex removed from an epilepsy surgery was also analyzed to determine dopamine 365

levels in non-cancerous brain tissue. These assays revealed dopamine expression in all cases 366

(Figure 4D). 367

In light of our previous data indicating that dopamine signaling is associated with cell state, we 368

next examined how cell state influences dopamine secretion. PDX GBM6 cells were cultured in 369

either 1% FBS or neurobasal media. Conditioned media were collected after 3, 14, and 21 days 370

and dopamine levels determined by HPLC-MS. We found that dopamine production was time 371

dependent, but not clearly linked to culture conditions (Figure 4E). Furthermore, it has been 372

established that hypoxia induction can influence the ability of stem cells to differentiate and 373

produce dopamine (Wang et al. 2013). We therefore analyzed dopamine levels from media in 374

which PDX GBM cells were growing exposed to either 20% normoxia or 1% hypoxia for 3 375

Page 18: Activation of dopamine receptor 2 (DRD2) prompts

17

hours. HPLC-MS revealed that hypoxia increase dopamine levels (Figure 4F), suggesting a 376

similar mechanism. 377

378

Activation of DRD2 induces expression of HIF proteins 379

To determine how DRD2 signaling may underlie the observed changes in self-renewing 380

capacity, we analyzed patient tumor samples from The Cancer Genome Atlas (TCGA). Patient 381

samples were stratified from lowest to highest expression of DRD2 and correlations were 382

determined for 12,042 genes by Pearson correlation coefficient analysis; coefficients >0.5 or <-383

0.5 and false discovery rate (FDR) <0.05 were used. Genes fitting these parameters were then 384

analyzed using Enrichr pathway analysis program. Our analysis revealed that DRD2 levels are 385

positively correlated with the expression of hypoxia inducible factors (HIF) 1α and HIF2α 386

pathways, as well as with the Reelin and ErbB4 signaling networks (Adjusted p-value= .03044) 387

(Figure 5A). 388

Hypoxia is well known to influence the phenotype of GBM cells, pushing them towards a more 389

GIC phenotype (Evans et al. 2004; Soeda et al. 2009). Further, we have previously shown that 390

GBM cells utilize HIF signaling to promote cellular plasticity following therapy (Lee et al. 391

2016). To investigate how DRD2 stimulation affects HIF signaling, PDX cells GBM39 and 392

GBM43 were treated with agonist under normal and hypoxic conditions. Immunoblot analysis 393

demonstrated a substantial, time-dependent increase in HIF1α levels in both cell lines following 394

activation of DRD2 signaling, regardless of oxygen availability (Figure 5B). 395

HIF protein levels are controlled both at the level of gene transcription and via hydroxylase-396

dependent degradation. In order to determine if DRD2’s activation of HIF was occurring at the 397

level of gene transcription or degradation, we analyzed the levels of PHD and VHL, key 398

Page 19: Activation of dopamine receptor 2 (DRD2) prompts

18

regulators of HIF stabilization, following treatment with the DRD2 agonists by western blot. 399

These assays revealed cell line specific changes in the HIF pathway. In GBM12, the expected 400

correlations were observed—elevated HIF levels matched decreased levels of both PHD and 401

VHL. However, in GBM39, the increased HIF expression actually correlated with elevated PHD 402

and VHL (Figure 5C). Notably, these results indicate that increases in HIF levels following 403

DRD2 activation occurs in both cell lines that are responsive and non-responsive in neurosphere 404

assays. This common shared HIF induction with differential functional outcomes suggests that 405

shared activation of HIF leads to unique downstream effects. 406

We next sought to determine if the increased GIC population following DRD2 activation was 407

dependent on the previously identified increase in HIF1α in the clinically relevant PDX model. 408

GBM 6 treated with HIF1α inhibitor after exposure to DRD2 agonist show loss of Sox2, CMYC, 409

and MMP2 expression (Figure 5D). 410

411

Activation of DRD2 induces alterations in expression of HIF-responsive genes and functional 412

changes in metabolic phenotype 413

HIF signaling can have profound effects on cancer cells, ranging from shifts in gene expression 414

to altered functional attributes. For a full transcriptome profile of the mechanisms governing 415

DRD2’s influence on GBM, we performed microarray analysis of gene expression in cells 416

treated with either vehicle control DMSO or 30nM DRD2 agonist for four days. We chose two 417

cell lines with distinct responses to dopamine in sphere-forming assays—neurosphere-responsive 418

GBM39 and non-responsive GBM12. To determine the changes that were specific to each cell 419

line, we utilized the online GeneVenn tool. Transcripts with a p value <.05 from each cell line, 420

comparing agonist and DMSO controls, were identified and overlap was analyzed by GeneVenn. 421

Page 20: Activation of dopamine receptor 2 (DRD2) prompts

19

We found that 3,479 increases in gene expression were specific to neurosphere-responsive 422

GBM39, while 1,917 increases were unique to neurosphere-non-responsive GBM12. These two 423

cell lines showed shared increases in only 287 genes (Figure 6A). Enrichr analysis of these cell 424

lines’ dopamine-induced transcriptomes revealed activation of unique signaling pathways 425

(Figure 6B). Given our result indicating a connection between HIF signaling and DRD2, we next 426

examined genes known to be regulated by HIF during canonical hypoxia responses. We therefore 427

performed GSEA analysis of only the hypoxia response gene-sets. This analysis confirmed a 428

subtype-dependent response to DRD2 activation. GBM39 cells did not activate canonical HIF 429

targets following agonist, whereas GBM12 did (Figure 6B). These results indicate a specific 430

change in gene expression following the agonist and suggest that these differences may underlie 431

the functional changes in sphere-formation. 432

While many genes are activated in response to HIF proteins, certain genes are known to be 433

activated via direct binding of HIFs to gene promoters at conserved regions termed HIF-434

responsive elements (HRE). To determine if the observed changes in gene expression occurred at 435

the level of actual HIF binding, we collected all the gene transcripts that were significantly 436

upregulated after treatment with the DRD2 agonist and known to have a promoter containing the 437

HRE (previously confirmed by ChIP-Seq(Schodel et al. 2011)). These sets of genes were then 438

analyzed using the Enrichr platform. Once again, we found that the two cell lines showed 439

drastically different responses to the agonist (Figure 6C). Treated GBM39 cells showed 440

increased levels of HIF-regulated genes, including glycolysis (p-value = 2.142E-7), whereas 441

GBM12 showed increased activation of the PI3K/AKT pathway (p-value = .001264) (Figure 442

6D). These results suggest that the differential effects of DRD2-induced activation of HIF in 443

classical GBM39 and proneural GBM12 depend on unique signaling mechanisms. 444

Page 21: Activation of dopamine receptor 2 (DRD2) prompts

20

Changes in gene expression, while informative, do not guarantee a functional alteration in 445

cellular behavior. We therefore sought to determine if activation of DRD2 does in fact lead to 446

GBM39-specific alterations in metabolism. First, we analyzed the uptake of glucose and fatty 447

acid via FACS, utilizing 2-NBDG, a well-established fluorescent analogue of glucose, and our 448

proprietary fatty-acid tagged quantum dots (Muroski et al. 2017). Only GBM39 showed altered 449

fatty acid uptake following DRD2 activation. However, all cells treated with the agonist showed 450

increases in glucose uptake (Extended Data Figure 6-1). To further characterize the effects of 451

DRD2 signaling on glucose metabolism, we performed extracellular flux analysis. We 452

discovered a differential effect predicted by each cell line’s behavior in neurosphere assays. 453

GBM39, of the classical subtype and responsive to the agonist in neurosphere assays, showed a 454

marked increase in glycolytic rate as measured by glucose stimulated extracellular acidification 455

(p-value = .00010) (ECAR) (Figure 6E). In contrast GBM 12, the non-responsive proneural 456

subtype, actually showed decreases in glycolysis following DRD2 agonist treatment (Figure 6F) 457

(p-value = .00011). 458

459

Discussion 460

In this study, we investigated how DRD2 activation contributes to the molecular and functional 461

phenotype of GBM. We demonstrated that therapeutic stress induced by anti-glioma 462

chemotherapy alters the epigenetic status of the DRD2 promoter and subsequently increases 463

DRD2 protein expression. Notably, this expression was elevated in GICs and could be induced 464

by GIC-promoting culture conditions. In classical GBM cells, DRD2 activation increased sphere 465

forming capacity, an indicator of GIC state and tumorgenicity, as well as the expression of 466

several key GIC markers. We further found that GBM cells themselves can generate dopamine in 467

Page 22: Activation of dopamine receptor 2 (DRD2) prompts

21

purified monocultures, and therefore exhibit autocrine/paracrine DRD2 activation capabilities. In 468

all lines tested, activation of DRD2 induced the expression of HIF proteins, regardless of oxygen 469

availability. Whereas the HIF induction by DRD2 activation was common to all cell lines tested, 470

the downstream functional changes varied. Classical GBM39 cells exhibited increased uptake of 471

glucose and glycolytic rate following DRD2 stimulation. In contrast, proneural GBM12 cells 472

showed no change in sphere-forming capacity. However, activation of DRD2 did induce 473

expression of canonical HIF-responsive genes and led to a reduction in their glycolytic rate. 474

Cells exposed to low oxygen conditions typically activate HIF and subsequently upregulate 475

glycolysis; the fact that proneural cells do not upregulate this metabolism, despite elevated HIF 476

levels, suggests a unique HIF regulatory mechanism. These results indicate a key role for DRD2 477

signaling in influencing GBM cellular plasticity, at the level of both gene expression and 478

functional attributes. 479

Induction of cellular plasticity, specifically in steering cells towards a treatment-resistant GIC 480

state, is a key problem in GBM and contributes to tumor recurrence (Safa et al. 2015; Olmez et 481

al. 2015; Dahan et al. 2014; Lee et al. 2016). Our data demonstrate that dopamine signaling plays 482

a role in this cellular conversion. Functionally, exposure to DRD2 agonist was sufficient to 483

increase sphere-forming capacity of PDX GBM cells. Given the concurrent increase in GIC 484

marker expression as well as engraftment efficiency, this treatment appears to induce the 485

conversion of GBM cells to a more stem-like state. 486

Our results indicate that activation of DRD2 induces unique downstream effects dependent on 487

the genetic subtype of cell line used. Even more interestingly, these unique changes in gene 488

expression and phenotypes occurred despite shared activation of a key signaling node—hypoxia 489

inducible factors. GBM39, of the classical tumor subtype, responds to the agonist with increased 490

Page 23: Activation of dopamine receptor 2 (DRD2) prompts

22

sphere-formation, expression of key GIC markers, altered gene expression related to metabolism, 491

and increased glycolytic rate. In contrast, GBM12, a proneural cell line, did not increase 492

neurosphere formation or expression of GIC markers, but did show induction of HIFs and altered 493

gene expression related to canonical HIF signaling. It remains an open question as to which other 494

signaling mechanisms when activated in these two cell lines enable common signaling nodes to 495

induce cell-line specific changes in gene expression and functional phenotype. The unique 496

genetic makeup of each cell line may provide an explanation of this phenomenon. Analysis of 497

patient sample gene expression and orthotopic xenograft tumor protein levels showed that 498

proneural tumors and GBM12 xenografts have somewhat elevated expression of DRD2 499

(Extended Data Figure 1-1), suggesting that this signaling axis may already be active. Further, 500

GBM39 is of the classical subtype and carries the EGFRvIII mutation, while GBM12 expresses 501

wildtype EGFR (Giannini et al. 2005). EGFR is connected to dopamine signaling in neural 502

development (Hoglinger et al. 2004); therefore, the fact that EGFR mutant cell lines respond 503

with increased sphere-formation suggests that increased EGFR activity may play a critical role in 504

the ability of DRD2 to alter cellular plasticity. 505

These differential responses to activation of the same receptor further highlight a key challenge 506

facing the neuro-oncology community—intratumoral heterogeneity. GBM tumors have been 507

shown to contain cells corresponding to each of the three subtypes within a single tumor 508

(Sottoriva et al. 2013; Patel et al. 2014). This wide range of cell types and diverse molecular 509

phenotypes currently limit the effective treatment of GBM. These data highlight the importance 510

of this heterogeneity, by suggesting a mechanism for the same molecular signal to exert unique 511

influences on different subpopulations of tumor cells. It is not hard to imagine that dopamine 512

Page 24: Activation of dopamine receptor 2 (DRD2) prompts

23

may generate multiple adaptive features, including elevated self-renewal capacity or 513

maintenance of the GIC-pool via increased glycolysis, and neo-angiogenesis in GBM cells. 514

These data illustrate that dopamine signaling exerts influence on global as well as HIF-dependent 515

gene expression. Further, our data demonstrate that these changes in gene transcription have a 516

functional effect, as dopamine signaling is capable of altering the metabolism of GBM cells. 517

GBM39 showed a highly elevated glycolytic rate following agonist treatment, while GBM12 518

cells had slightly reduced rates of glycolysis. How this increase in glucose uptake and glycolytic 519

rate relates to induction of the GIC state, however, remains an open question. Several groups 520

have recently demonstrated that altered metabolic phenotypes influence the acquisition of a stem 521

cell phenotype in cancer cells (Currie et al. 2013; Mao et al. 2013); it is not outside the realm of 522

possibility that a similar effect is at play here. Further research will delineate the precise 523

connection between elevated glucose uptake and glycolytic rate and altered cell state. 524

It should be noted that a recent report suggests that dopamine may actually inhibit growth of 525

glioma. Lan and colleagues showed that dopamine reduced growth of U87 and U251 cells 526

growing as subcutaneous xenografts (Lan et al. 2017). While difference in cell lines (U87 v. 527

PDX) as well as the subcutaneous placement of tumors may account for this difference, this 528

result suggests a dose-dependent effect of dopamine. Critically, our experiments were performed 529

with doses of DRD2 agonist between 20 and 30nM, while Lan et al. performed experiments with 530

doses of dopamine between 10-25μΜ. Dopamine receptor signaling is highly sensitive to 531

concentration of ligands and antagonists, which complicates understanding its role in GBM 532

(reviewed (Caragher et al. 2017)). It is possible that the influence of dopamine signaling is dose-533

dependent. Further, this work highlights the complex nature of dopamine’s multiple receptors. 534

Page 25: Activation of dopamine receptor 2 (DRD2) prompts

24

Adding a pan dopamine receptor agonist, as they did, will activate all five receptors, which have 535

unique downstream signaling pathways. 536

Our data also show that GBM cells can synthesize and secrete dopamine, an ability assumed to 537

be the sole province of neurons. However, there is mounting evidence that GBM cells have the 538

ability to adopt certain neuronal functions. Osswald et al. demonstrated that GBM cells utilize 539

GAP43, neuronal outgrowth cone protein, to form networks (Osswald et al. 2015). Further, it has 540

been shown that GBM cells synthesize and secrete glutamate, an excitatory neurotransmitter, and 541

alter cortical activity (Buckingham et al. 2011). Developmental neurobiology also provides some 542

clues as to how these cells can attain a neuron-specific behavior. Neural stem cells engrafted into 543

the striatum of mice can spontaneously attain dopaminergic traits, suggesting that secreted 544

factors in the brain can induce the formation of dopamine secreting cells (Yang et al. 2002). 545

Given the highly similar expression profile of NSCs and GSCs, a similar mechanism may induce 546

the formation of a dopamine-producing population of glioma cells. Finally, a recent report 547

utilizing TCGA datasets indicated that high TH expression correlates with reduced median 548

survival (Dolma et al. 2016). Critically, this data analyzed mRNA expression in GBM cells 549

themselves, not in surrounding brain cells. Therefore, the survival difference relies on how TH 550

functions in the tumor. 551

In short, GBM cells exhibit a variety of neuron-like properties, including the ability to synthesize 552

and secrete dopamine. Our in vitro data indicate that the GBM cells are synthesizing dopamine 553

rather than taking up dopamine from the surrounding dopamine neurons and storing it. Our 554

results clearly fit within this emerging theory that GBM tumors may display neuron-specific 555

behaviors. More research is needed to delineate the specific subpopulations of GBM tumors that 556

secrete dopamine and the mechanisms governing this process. This result, combined with the 557

Page 26: Activation of dopamine receptor 2 (DRD2) prompts

25

evidence that DRD2 signaling promotes a shift to the GIC state, suggests that tumors utilize 558

dopamine as a mechanism to control their microenvironment and promote a pro-growth, pro-559

stemness milieu. 560

It could be argued that even if GBM cells do utilize dopamine in an autocrine manner, they must 561

also rely on ambient dopamine in the brain microenvironment to induce major signaling changes. 562

It remains possible that non-tumor dopamine also influences GBM. It has already been shown 563

that proteins from the synaptic cleft of nearby neurons can influence glioma growth (Venkatesh 564

et al. 2015). We therefore cannot rule out the possibility that our tumor sample data represent 565

cells that have absorbed dopamine rather than produced it (Figure 4). Indeed, TCGA indicates 566

that GBM cells express the dopamine transporter (Extended Data Figure 4-1). In addition, it has 567

been shown that patients with Parkinson’s Disease, characterized by the loss of dopaminergic 568

neurons, have a lower incidence of high grade gliomas (Diamandis et al. 2009). Further, studies 569

of tumor localization indicate that GBM tumors preferentially localize to areas of dopamine 570

innervation (Larjavaara et al. 2007). We do not exclude or reject the possibility that ambient 571

dopamine influences tumor behavior, with two key caveats. First, given that only specific 572

portions of the brain contain dopaminergic neurons (Ungerstedt 1971) and that GBM tumor 573

growth often disrupts the normal neural circuitry in their area, it cannot be guaranteed that 574

dopamine from the microenvironment is widely available for every tumor. Second, the three-575

dimensional shape of the tumor is likely to limit perfusion of certain molecules from distal 576

sources into the cleft. As such, ambient dopamine released from neurons may also influence 577

signaling in GBM tumors. 578

Finally, it remains a lingering question the extent to which the effects of dopamine signaling 579

observed here are GBM-specific or represent a recapitulation of the behavior of normal neural 580

Page 27: Activation of dopamine receptor 2 (DRD2) prompts

26

stem cells (NSCs) and brain progenitor cells. It has been demonstrated, for example, that NSCs 581

in the subventricular zone respond to dopamine with increased proliferation(Baker, Baker, and 582

Hagg 2004; O'Keeffe et al. 2009; Winner et al. 2009). Activation of dopamine receptors on 583

progenitors in the developing brain has also been shown to expand neuronal populations and 584

neurite outgrowth (Yoon and Biak 2013), and alter cell cycle status (Ohtani et al. 2003). Finally, 585

dopamine signaling in oligodendrocyte precursors (OPCs) expands the pluripotent population 586

(Kimoto et al. 2011). These studies suggest that GBM cells have hijacked an existing role for 587

dopamine in brain development for their own gains. The connection of metabolism, however, 588

was not identified in these studies or other relevant literature. Further research will undoubtedly 589

expand on this interesting connection. 590

In sum, our data indicate that GBM cells respond to DRD2 activation at the level of both gene 591

expression and functional phenotype and that specific inactivation of DRD2 itself is required to 592

provide therapeutic benefit (see Figure 3 and Extended Data Figure 3-1). Further research will 593

shed light on the dynamics of tumor-derived dopamine and mechanisms influencing key factors 594

like rate of production and secretion, interaction with non-cancerous cells in the environment, 595

and the effects of tumor-derived dopamine on therapy resistance. Novel creation of specific 596

dopamine receptor inhibitors may provide better clinical relevance towards treatment of tumor 597

types that rely on dopamine signaling dynamics. Overall, this study highlights a novel 598

mechanism by which GBM cultivates a supportive microenvironmental niche to colonize the 599

CNS. It also suggests that GBM tumors possess a remarkable ability to hijack cellular 600

mechanisms previously thought to be the exclusive provenance of neurons. Given the growing 601

chorus of studies indicating that dopamine antagonists inhibit GBM growth (Kang et al. 2017; 602

Karpel-Massler et al. 2015; Shin et al. 2006), these results provide critical context for the source 603

Page 28: Activation of dopamine receptor 2 (DRD2) prompts

27

of tumor-influencing dopamine and the mechanism underlying dopamine’s role in 604

gliomagenesis. 605

606

607

608

609

610

611

612

613

614

615

616

617

618

619

620

References: 621

Alifieris, C., and D. T. Trafalis. 2015. 'Glioblastoma multiforme: Pathogenesis and treatment', Pharmacol 622

Ther, 152: 63-82. 623

Baker, S. A., K. A. Baker, and T. Hagg. 2004. 'Dopaminergic nigrostriatal projections regulate neural 624

precursor proliferation in the adult mouse subventricular zone', Eur J Neurosci, 20: 575-9. 625

Beaulieu, J. M., and R. R. Gainetdinov. 2011. 'The physiology, signaling, and pharmacology of dopamine 626

receptors', Pharmacol Rev, 63: 182-217. 627

Page 29: Activation of dopamine receptor 2 (DRD2) prompts

28

Bowman, R. L., Q. Wang, A. Carro, R. G. Verhaak, and M. Squatrito. 2017. 'GlioVis data portal for 628

visualization and analysis of brain tumor expression datasets', Neuro Oncol, 19: 139-41. 629

Buckingham, S. C., S. L. Campbell, B. R. Haas, V. Montana, S. Robel, T. Ogunrinu, and H. Sontheimer. 630

2011. 'Glutamate release by primary brain tumors induces epileptic activity', Nat Med, 17: 1269-631

74. 632

Caragher, S. P., R. R. Hall, 3rd, R. Ahsan, and A. U. Ahmed. 2017. 'Monoamines in Glioblastoma: 633

complex biology with therapeutic potential', Neuro Oncol. 634

Charles, N. A., E. C. Holland, R. Gilbertson, R. Glass, and H. Kettenmann. 2011. 'The brain tumor 635

microenvironment', Glia, 59: 1169-80. 636

Currie, E., A. Schulze, R. Zechner, T. C. Walther, and R. V. Farese, Jr. 2013. 'Cellular fatty acid 637

metabolism and cancer', Cell Metab, 18: 153-61. 638

Dahan, P., J. Martinez Gala, C. Delmas, S. Monferran, L. Malric, D. Zentkowski, V. Lubrano, C. Toulas, 639

E. Cohen-Jonathan Moyal, and A. Lemarie. 2014. 'Ionizing radiations sustain glioblastoma cell 640

dedifferentiation to a stem-like phenotype through survivin: possible involvement in 641

radioresistance', Cell Death Dis, 5: e1543. 642

Diamandis, P., A. G. Sacher, M. Tyers, and P. B. Dirks. 2009. 'New drugs for brain tumors? Insights from 643

chemical probing of neural stem cells', Med Hypotheses, 72: 683-7. 644

Dolma, S., H. J. Selvadurai, X. Lan, L. Lee, M. Kushida, V. Voisin, H. Whetstone, M. So, T. Aviv, N. 645

Park, X. Zhu, C. Xu, R. Head, K. J. Rowland, M. Bernstein, I. D. Clarke, G. Bader, L. 646

Harrington, J. H. Brumell, M. Tyers, and P. B. Dirks. 2016. 'Inhibition of Dopamine Receptor D4 647

Impedes Autophagic Flux, Proliferation, and Survival of Glioblastoma Stem Cells', Cancer Cell, 648

29: 859-73. 649

Evans, S. M., K. D. Judy, I. Dunphy, W. T. Jenkins, W. T. Hwang, P. T. Nelson, R. A. Lustig, K. Jenkins, 650

D. P. Magarelli, S. M. Hahn, R. A. Collins, M. S. Grady, and C. J. Koch. 2004. 'Hypoxia is 651

important in the biology and aggression of human glial brain tumors', Clin Cancer Res, 10: 8177-652

84. 653

Page 30: Activation of dopamine receptor 2 (DRD2) prompts

29

Floresco, S. B., A. R. West, B. Ash, H. Moore, and A. A. Grace. 2003. 'Afferent modulation of dopamine 654

neuron firing differentially regulates tonic and phasic dopamine transmission', Nat Neurosci, 6: 655

968-73. 656

Galli, R., E. Binda, U. Orfanelli, B. Cipelletti, A. Gritti, S. De Vitis, R. Fiocco, C. Foroni, F. Dimeco, and 657

A. Vescovi. 2004. 'Isolation and characterization of tumorigenic, stem-like neural precursors from 658

human glioblastoma', Cancer Res, 64: 7011-21. 659

Giannini, C., J. N. Sarkaria, A. Saito, J. H. Uhm, E. Galanis, B. L. Carlson, M. A. Schroeder, and C. D. 660

James. 2005. 'Patient tumor EGFR and PDGFRA gene amplifications retained in an invasive 661

intracranial xenograft model of glioblastoma multiforme', Neuro Oncol, 7: 164-76. 662

Hardee, M. E., A. E. Marciscano, C. M. Medina-Ramirez, D. Zagzag, A. Narayana, S. M. Lonning, and 663

M. H. Barcellos-Hoff. 2012. 'Resistance of glioblastoma-initiating cells to radiation mediated by 664

the tumor microenvironment can be abolished by inhibiting transforming growth factor-beta', 665

Cancer Res, 72: 4119-29. 666

Heddleston, J. M., Z. Li, R. E. McLendon, A. B. Hjelmeland, and J. N. Rich. 2009. 'The hypoxic 667

microenvironment maintains glioblastoma stem cells and promotes reprogramming towards a 668

cancer stem cell phenotype', Cell Cycle, 8: 3274-84. 669

Hjelmeland, A. B., Q. Wu, J. M. Heddleston, G. S. Choudhary, J. MacSwords, J. D. Lathia, R. 670

McLendon, D. Lindner, A. Sloan, and J. N. Rich. 2011. 'Acidic stress promotes a glioma stem 671

cell phenotype', Cell Death Differ, 18: 829-40. 672

Hodgson, J. G., R. F. Yeh, A. Ray, N. J. Wang, I. Smirnov, M. Yu, S. Hariono, J. Silber, H. S. Feiler, J. 673

W. Gray, P. T. Spellman, S. R. Vandenberg, M. S. Berger, and C. D. James. 2009. 'Comparative 674

analyses of gene copy number and mRNA expression in glioblastoma multiforme tumors and 675

xenografts', Neuro Oncol, 11: 477-87. 676

Hoglinger, G. U., P. Rizk, M. P. Muriel, C. Duyckaerts, W. H. Oertel, I. Caille, and E. C. Hirsch. 2004. 677

'Dopamine depletion impairs precursor cell proliferation in Parkinson disease', Nat Neurosci, 7: 678

726-35. 679

Page 31: Activation of dopamine receptor 2 (DRD2) prompts

30

Huang, M., J. J. Panos, S. Kwon, Y. Oyamada, L. Rajagopal, and H. Y. Meltzer. 2014. 'Comparative 680

effect of lurasidone and blonanserin on cortical glutamate, dopamine, and acetylcholine efflux: 681

role of relative serotonin (5-HT)2A and DA D2 antagonism and 5-HT1A partial agonism', J 682

Neurochem, 128: 938-49. 683

Kang, S., J. Hong, J. M. Lee, H. E. Moon, B. Jeon, J. Choi, N. A. Yoon, S. H. Paek, E. J. Roh, C. J. Lee, 684

and S. S. Kang. 2017. 'Trifluoperazine, a Well-Known Antipsychotic, Inhibits Glioblastoma 685

Invasion by Binding to Calmodulin and Disinhibiting Calcium Release Channel IP3R', Mol 686

Cancer Ther, 16: 217-27. 687

Karpel-Massler, G., R. E. Kast, M. A. Westhoff, A. Dwucet, N. Welscher, L. Nonnenmacher, M. Hlavac, 688

M. D. Siegelin, C. R. Wirtz, K. M. Debatin, and M. E. Halatsch. 2015. 'Olanzapine inhibits 689

proliferation, migration and anchorage-independent growth in human glioblastoma cell lines and 690

enhances temozolomide's antiproliferative effect', J Neurooncol, 122: 21-33. 691

Keefe, K. A., M. J. Zigmond, and E. D. Abercrombie. 1993. 'In vivo regulation of extracellular dopamine 692

in the neostriatum: influence of impulse activity and local excitatory amino acids', J Neural 693

Transm Gen Sect, 91: 223-40. 694

Kimoto, S., A. Okuda, M. Toritsuka, T. Yamauchi, M. Makinodan, H. Okuda, K. Tatsumi, Y. Nakamura, 695

A. Wanaka, and T. Kishimoto. 2011. 'Olanzapine stimulates proliferation but inhibits 696

differentiation in rat oligodendrocyte precursor cell cultures', Prog Neuropsychopharmacol Biol 697

Psychiatry, 35: 1950-6. 698

Lammel, S., B. K. Lim, and R. C. Malenka. 2014. 'Reward and aversion in a heterogeneous midbrain 699

dopamine system', Neuropharmacology, 76 Pt B: 351-9. 700

Lan, Y. L., X. Wang, J. S. Xing, Z. L. Yu, J. C. Lou, X. C. Ma, and B. Zhang. 2017. 'Anti-cancer effects 701

of dopamine in human glioma: involvement of mitochondrial apoptotic and anti-inflammatory 702

pathways', Oncotarget, 8: 88488-500. 703

Larjavaara, S., R. Mantyla, T. Salminen, H. Haapasalo, J. Raitanen, J. Jaaskelainen, and A. Auvinen. 704

2007. 'Incidence of gliomas by anatomic location', Neuro Oncol, 9: 319-25. 705

Page 32: Activation of dopamine receptor 2 (DRD2) prompts

31

Lee, G., B. Auffinger, D. Guo, T. Hasan, M. Deheeger, A. L. Tobias, J. Y. Kim, F. Atashi, L. Zhang, M. 706

S. Lesniak, J. C. David, and A. U. Ahmed. 2016. 'Dedifferentiation of Glioma Cells to Glioma 707

Stem-like Cells By Therapeutic Stress-induced HIF Signaling in the Recurrent GBM Model', Mol 708

Cancer Ther, 15: 3064-76. 709

Li, J., S. Zhu, D. Kozono, K. Ng, D. Futalan, Y. Shen, J. C. Akers, T. Steed, D. Kushwaha, M. Schlabach, 710

B. S. Carter, C. H. Kwon, F. Furnari, W. Cavenee, S. Elledge, and C. C. Chen. 2014. 'Genome-711

wide shRNA screen revealed integrated mitogenic signaling between dopamine receptor D2 712

(DRD2) and epidermal growth factor receptor (EGFR) in glioblastoma', Oncotarget, 5: 882-93. 713

Mao, P., K. Joshi, J. Li, S. H. Kim, P. Li, L. Santana-Santos, S. Luthra, U. R. Chandran, P. V. Benos, L. 714

Smith, M. Wang, B. Hu, S. Y. Cheng, R. W. Sobol, and I. Nakano. 2013. 'Mesenchymal glioma 715

stem cells are maintained by activated glycolytic metabolism involving aldehyde dehydrogenase 716

1A3', Proc Natl Acad Sci U S A, 110: 8644-9. 717

Mukherjee, B., B. McEllin, C. V. Camacho, N. Tomimatsu, S. Sirasanagandala, S. Nannepaga, K. J. 718

Hatanpaa, B. Mickey, C. Madden, E. Maher, D. A. Boothman, F. Furnari, W. K. Cavenee, R. M. 719

Bachoo, and S. Burma. 2009. 'EGFRvIII and DNA double-strand break repair: a molecular 720

mechanism for radioresistance in glioblastoma', Cancer Res, 69: 4252-9. 721

Muroski, M. E., J. Miska, A. L. Chang, P. Zhang, A. Rashidi, H. Moore, A. Lopez-Rosas, Y. Han, and M. 722

S. Lesniak. 2017. 'Fatty Acid Uptake in T Cell Subsets Using a Quantum Dot Fatty Acid 723

Conjugate', Sci Rep, 7: 5790. 724

O'Keeffe, G. C., P. Tyers, D. Aarsland, J. W. Dalley, R. A. Barker, and M. A. Caldwell. 2009. 725

'Dopamine-induced proliferation of adult neural precursor cells in the mammalian subventricular 726

zone is mediated through EGF', Proc Natl Acad Sci U S A, 106: 8754-9. 727

Ohtani, N., T. Goto, C. Waeber, and P. G. Bhide. 2003. 'Dopamine modulates cell cycle in the lateral 728

ganglionic eminence', J Neurosci, 23: 2840-50. 729

Page 33: Activation of dopamine receptor 2 (DRD2) prompts

32

Olmez, I., W. Shen, H. McDonald, and B. Ozpolat. 2015. 'Dedifferentiation of patient-derived 730

glioblastoma multiforme cell lines results in a cancer stem cell-like state with mitogen-731

independent growth', J Cell Mol Med, 19: 1262-72. 732

Osswald, M., E. Jung, F. Sahm, G. Solecki, V. Venkataramani, J. Blaes, S. Weil, H. Horstmann, B. 733

Wiestler, M. Syed, L. Huang, M. Ratliff, K. Karimian Jazi, F. T. Kurz, T. Schmenger, D. Lemke, 734

M. Gommel, M. Pauli, Y. Liao, P. Haring, S. Pusch, V. Herl, C. Steinhauser, D. Krunic, M. 735

Jarahian, H. Miletic, A. S. Berghoff, O. Griesbeck, G. Kalamakis, O. Garaschuk, M. Preusser, S. 736

Weiss, H. Liu, S. Heiland, M. Platten, P. E. Huber, T. Kuner, A. von Deimling, W. Wick, and F. 737

Winkler. 2015. 'Brain tumour cells interconnect to a functional and resistant network', Nature, 738

528: 93-8. 739

Patel, A. P., I. Tirosh, J. J. Trombetta, A. K. Shalek, S. M. Gillespie, H. Wakimoto, D. P. Cahill, B. V. 740

Nahed, W. T. Curry, R. L. Martuza, D. N. Louis, O. Rozenblatt-Rosen, M. L. Suva, A. Regev, 741

and B. E. Bernstein. 2014. 'Single-cell RNA-seq highlights intratumoral heterogeneity in primary 742

glioblastoma', Science, 344: 1396-401. 743

Safa, A. R., M. R. Saadatzadeh, A. A. Cohen-Gadol, K. E. Pollok, and K. Bijangi-Vishehsaraei. 2015. 744

'Glioblastoma stem cells (GSCs) epigenetic plasticity and interconversion between differentiated 745

non-GSCs and GSCs', Genes Dis, 2: 152-63. 746

Schodel, J., S. Oikonomopoulos, J. Ragoussis, C. W. Pugh, P. J. Ratcliffe, and D. R. Mole. 2011. 'High-747

resolution genome-wide mapping of HIF-binding sites by ChIP-seq', Blood, 117: e207-17. 748

Schunemann, D. P., I. Grivicich, A. Regner, L. F. Leal, D. R. de Araujo, G. P. Jotz, C. A. Fedrigo, D. 749

Simon, and A. B. da Rocha. 2010. 'Glutamate promotes cell growth by EGFR signaling on U-750

87MG human glioblastoma cell line', Pathol Oncol Res, 16: 285-93. 751

Shin, S. Y., B. H. Choi, J. Ko, S. H. Kim, Y. S. Kim, and Y. H. Lee. 2006. 'Clozapine, a neuroleptic 752

agent, inhibits Akt by counteracting Ca2+/calmodulin in PTEN-negative U-87MG human 753

glioblastoma cells', Cell Signal, 18: 1876-86. 754

Page 34: Activation of dopamine receptor 2 (DRD2) prompts

33

Singh, S. K., C. Hawkins, I. D. Clarke, J. A. Squire, J. Bayani, T. Hide, R. M. Henkelman, M. D. 755

Cusimano, and P. B. Dirks. 2004. 'Identification of human brain tumour initiating cells', Nature, 756

432: 396-401. 757

Snuderl, Matija, Ladan Fazlollahi, Long P. Le, Mai Nitta, Boryana H. Zhelyazkova, Christian J. 758

Davidson, Sara Akhavanfard, Daniel P. Cahill, Kenneth D. Aldape, Rebecca A. Betensky, David 759

N. Louis, and John A. Iafrate. 2011. 'Mosaic Amplification of Multiple Receptor Tyrosine Kinase 760

Genes in Glioblastoma', Cancer Cell, 20: 810-17. 761

Soeda, A., M. Park, D. Lee, A. Mintz, A. Androutsellis-Theotokis, R. D. McKay, J. Engh, T. Iwama, T. 762

Kunisada, A. B. Kassam, I. F. Pollack, and D. M. Park. 2009. 'Hypoxia promotes expansion of 763

the CD133-positive glioma stem cells through activation of HIF-1alpha', Oncogene, 28: 3949-59. 764

Sottoriva, A., I. Spiteri, S. G. Piccirillo, A. Touloumis, V. P. Collins, J. C. Marioni, C. Curtis, C. Watts, 765

and S. Tavare. 2013. 'Intratumor heterogeneity in human glioblastoma reflects cancer 766

evolutionary dynamics', Proc Natl Acad Sci U S A, 110: 4009-14. 767

Stupp, R., S. Taillibert, A. Kanner, W. Read, D. Steinberg, B. Lhermitte, S. Toms, A. Idbaih, M. S. 768

Ahluwalia, K. Fink, F. Di Meco, F. Lieberman, J. J. Zhu, G. Stragliotto, D. Tran, S. Brem, A. 769

Hottinger, E. D. Kirson, G. Lavy-Shahaf, U. Weinberg, C. Y. Kim, S. H. Paek, G. Nicholas, J. 770

Bruna, H. Hirte, M. Weller, Y. Palti, M. E. Hegi, and Z. Ram. 2017. 'Effect of Tumor-Treating 771

Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in 772

Patients With Glioblastoma: A Randomized Clinical Trial', JAMA, 318: 2306-16. 773

Szerlip, N. J., A. Pedraza, D. Chakravarty, M. Azim, J. McGuire, Y. Fang, T. Ozawa, E. C. Holland, J. T. 774

Huse, S. Jhanwar, M. A. Leversha, T. Mikkelsen, and C. W. Brennan. 2012. 'Intratumoral 775

heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma 776

defines subpopulations with distinct growth factor response', Proc Natl Acad Sci U S A, 109: 777

3041-6. 778

Page 35: Activation of dopamine receptor 2 (DRD2) prompts

34

Tamura, K., M. Aoyagi, N. Ando, T. Ogishima, H. Wakimoto, M. Yamamoto, and K. Ohno. 2013. 779

'Expansion of CD133-positive glioma cells in recurrent de novo glioblastomas after radiotherapy 780

and chemotherapy', J Neurosurg, 119: 1145-55. 781

Ungerstedt, U. 1971. 'Stereotaxic mapping of the monoamine pathways in the rat brain', Acta Physiol 782

Scand Suppl, 367: 1-48. 783

Venkatesh, H. S., T. B. Johung, V. Caretti, A. Noll, Y. Tang, S. Nagaraja, E. M. Gibson, C. W. Mount, J. 784

Polepalli, S. S. Mitra, P. J. Woo, R. C. Malenka, H. Vogel, M. Bredel, P. Mallick, and M. Monje. 785

2015. 'Neuronal Activity Promotes Glioma Growth through Neuroligin-3 Secretion', Cell, 161: 786

803-16. 787

Venkatesh, H. S., L. T. Tam, P. J. Woo, J. Lennon, S. Nagaraja, S. M. Gillespie, J. Ni, D. Y. Duveau, P. J. 788

Morris, J. J. Zhao, C. J. Thomas, and M. Monje. 2017. 'Targeting neuronal activity-regulated 789

neuroligin-3 dependency in high-grade glioma', Nature, 549: 533-37. 790

Verhaak, R. G., K. A. Hoadley, E. Purdom, V. Wang, Y. Qi, M. D. Wilkerson, C. R. Miller, L. Ding, T. 791

Golub, J. P. Mesirov, G. Alexe, M. Lawrence, M. O'Kelly, P. Tamayo, B. A. Weir, S. Gabriel, W. 792

Winckler, S. Gupta, L. Jakkula, H. S. Feiler, J. G. Hodgson, C. D. James, J. N. Sarkaria, C. 793

Brennan, A. Kahn, P. T. Spellman, R. K. Wilson, T. P. Speed, J. W. Gray, M. Meyerson, G. Getz, 794

C. M. Perou, D. N. Hayes, and Network Cancer Genome Atlas Research. 2010. 'Integrated 795

genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by 796

abnormalities in PDGFRA, IDH1, EGFR, and NF1', Cancer Cell, 17: 98-110. 797

Villa, G. R., J. J. Hulce, C. Zanca, J. Bi, S. Ikegami, G. L. Cahill, Y. Gu, K. M. Lum, K. Masui, H. Yang, 798

X. Rong, C. Hong, K. M. Turner, F. Liu, G. C. Hon, D. Jenkins, M. Martini, A. M. Armando, O. 799

Quehenberger, T. F. Cloughesy, F. B. Furnari, W. K. Cavenee, P. Tontonoz, T. C. Gahman, A. K. 800

Shiau, B. F. Cravatt, and P. S. Mischel. 2016. 'An LXR-Cholesterol Axis Creates a Metabolic Co-801

Dependency for Brain Cancers', Cancer Cell, 30: 683-93. 802

Page 36: Activation of dopamine receptor 2 (DRD2) prompts

35

Wang, Y., J. Yang, H. Li, X. Wang, L. Zhu, M. Fan, and X. Wang. 2013. 'Hypoxia promotes 803

dopaminergic differentiation of mesenchymal stem cells and shows benefits for transplantation in 804

a rat model of Parkinson's disease', PLoS One, 8: e54296. 805

Winkler, F., S. V. Kozin, R. T. Tong, S. S. Chae, M. F. Booth, I. Garkavtsev, L. Xu, D. J. Hicklin, D. 806

Fukumura, E. di Tomaso, L. L. Munn, and R. K. Jain. 2004. 'Kinetics of vascular normalization 807

by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, 808

angiopoietin-1, and matrix metalloproteinases', Cancer Cell, 6: 553-63. 809

Winner, B., P. Desplats, C. Hagl, J. Klucken, R. Aigner, S. Ploetz, J. Laemke, A. Karl, L. Aigner, E. 810

Masliah, E. Buerger, and J. Winkler. 2009. 'Dopamine receptor activation promotes adult 811

neurogenesis in an acute Parkinson model', Exp Neurol, 219: 543-52. 812

Yang, M., N. D. Stull, M. A. Berk, E. Y. Snyder, and L. Iacovitti. 2002. 'Neural stem cells spontaneously 813

express dopaminergic traits after transplantation into the intact or 6-hydroxydopamine-lesioned 814

rat', Exp Neurol, 177: 50-60. 815

Yoon, S., and J. H. Baik. 2013. 'Dopamine D2 receptor-mediated epidermal growth factor receptor 816

transactivation through a disintegrin and metalloprotease regulates dopaminergic neuron 817

development via extracellular signal-related kinase activation', J Biol Chem, 288: 28435-46. 818

819

820 821 822 823 824 825 826 827 828 829 830

Page 37: Activation of dopamine receptor 2 (DRD2) prompts

36

Figure 1: Chemotherapeutic stress induces DRD2 expression in the patient derived xenograft 831

glioma lines. (A/B) H3K27ac and H3K27me3 marks distribution around the transcription start 832

sites of dopamine receptors following therapy. Chromatin immunoprecipitation with massively 833

parallel DNA sequencing (ChIP-Seq) was performed for the open chromatin state histone 3 834

lysine 27 (H3K27) acetylation in PDX line GBM43 after 4 days of single exposure to 835

temozolomide (TMZ, 50μM), or treated with one fractionated dose of radiotherapy (2 Gy). 836

DMSO was used as a vehicle control. The red box in DRD2 highlights a key residue near the 837

promoter that uniquely exhibited increased H3K27ac following TMZ treatment. (MACS2 Peak 838

Score: 45.0, fold enrichment: 4.023, P-value < 0.0001). Table summarizes statistical information 839

for each of the 5 DRDs. Extended data included in Figure 1-1 also shows peak calling of all 840

monoamine receptors. (C) Western blot analysis of dopamine receptors expression in PDX cells 841

treated with TMZ over 8 days. (D) Fluorescence activated cell sorting analysis (FACS) analysis 842

of DRD2 on different subtype of PDX cells treated with vehicle control DMSO or TMZ (50μM). 843

Extended data figure 1-2 includes analysis of all 5 dopamine receptors demonstrating elevated 844

DRD2 expression in human samples of both sexes. Bars represent means from three independent 845

experiments and error bars show the standard deviation (SD). Student T-tests were performed 846

for each day separately. *P<.05, **P<.01, ***P<.001. 847

848 849 Figure 2: Glioma initiating cells preferentially express DRD2 and specifically respond to 850

receptor activation with hyperpolarization. (A) Two different subtypes of PDX lines freshly 851

isolated from animals were cultured in the tumor sphere maintenance media (neurobasal media 852

containing EGF and FGF) or differentiation media (1% fetal bovine serum) and subjected to 853

immunoblot analysis for the expression of DRD2 and various GIC markers. (B) PDX cells were 854

Page 38: Activation of dopamine receptor 2 (DRD2) prompts

37

subjected to FACS analysis for GIC marker CD133 and DRD2. Using controls, we established 855

gates for CD133+ and CD133- populations. We then quantified the mean fluorescence intensity 856

(MFI) for DRD2 expression in each of these populations. (C) Human glioma U251 cells 857

expressing red fluorescent protein (RFP) under the control of the CD133 promoter were patch-858

clamped and electrophysiological recordings were taken during exposure to highly selective 859

DRD2 agonist. Puffing DRD2 agonist (30nM, yellow) onto fluorescently labeled GIC (brightly 860

labeled cell left panel, red membrane potential trace middle panel) induces a more robust 861

hyperpolarization of the membrane potential (mV) than non-GICs exposed to the same agonist 862

(nsGBM, non-labeled cell left panel, blue trace middle panel), or versus puffing vehicle control 863

(n=4 each). Graph: DRD2 agonist hyperpolarizes GIC more strongly than nsGBM cells (right 864

panel). (D) Electrophysiological recordings of PDX cells confirms that only certain cells respond 865

to the agonist with hyperpolarization. Bars represent means from three independent experiments 866

and error bars show the SD. Student T-tests were performed for each day separately. *P<.05, 867

**P<.01, ***P<.001. 868

869 870 Figure 3: Activation of DRD2 signaling increases the self-renewal capacity of GBM cells. (A) 871

PDX GBM cells treated with either DMSO or TMZ were sorted based on DRD2 expression and 872

neurosphere assays were performed. Using the extreme limiting dilution assay algorithm, 873

frequency of GICs was calculated (DMSO, DRD2+ 1/458 and DRD2- 1/2091, p<.001; TMZ, 874

DRD+ 1/196 and DRD2- 1/515, p<.001). (B) In vivo engraftment of 500 cells sorted on DRD2 + 875

or – expression demonstrates DRD2+ cells engraft more efficiently. Extended data figure 3-1 876

includes more images of engrafted tumors from other mice. PDX lines treated with dopamine 877

agonist demonstrate elevation in key stem cell genes via western blot. Extended data figure 3-1 878

Page 39: Activation of dopamine receptor 2 (DRD2) prompts

38

includes western blots from other PDX lines demonstrating similar trends. (C) PDX GBM cells 879

were treated with either DRD2 agonist (30nM) or equimolar vehicle control DMSO and plated 880

for neurosphere assays. Using the extreme limiting dilution assay algorithm, frequency of GICs 881

was calculated (GBM6, DMSO stem cells 1/62 and agonist stem cells 1/29.2 (p<.01); GBM39, 882

DMSO stem cells 1/181.9 and agonist stem cells 1/87.8 (p<.05); GBM5, DMSO stem cells 883

1/118.9 and agonist stem cells 1/67.4 (p<.05). Extended data figure 3-1 includes a non-884

responding GBM12 neurosphere assay. (D) Two PDX lines were treated with either 885

chlorpromazine (CPM, 1μM) or equimolar DMSO and plated for neurosphere assays as above. 886

Fraction Non-responding graph is shown for GBM 39. Difference between groups was 887

determined by score test of heterogeneity. Error bars represent upper and lower limit of 95% 888

confidence interval computed for stem cell frequency. (E) Viral knockout of DRD2 using two 889

different SH-RNA constructs in GBM 43 demonstrates reduced stem cell frequency. Student T-890

Tests were performed for each comparison. ****P<0.0001. 891

892 893 894 Figure 4: GBM cells synthesize and secrete dopamine. (A) Western blot analysis of tyrosine 895

hydroxylase (TH) expression, the rate-limiting enzyme in dopamine synthesis. Beta-actin was 896

used as a loading control. (B) PDX cells were implanted intracranially into athymic nude mice. 897

Upon sickness from tumor burden, mice were sacrificed and whole brains extracted. FACS 898

analysis of human leukocyte antigen positive tumor cells demonstrates that GBM tumors 899

continue to express TH in vivo. Each dot represents individual mice; FACS were run in technical 900

triplicates. In set scatter plots are representative for FACS results in each cell line tested. (C) 901

Conditioned media from PDX GBM cells growing in purified monocultures were analyzed by 902

high performance liquid chromatography with mass spectrometry (HPLC-MS) for dopamine. 903

Page 40: Activation of dopamine receptor 2 (DRD2) prompts

39

Unconditioned DMEM media with and without 1% FBS was used as a control. (D) HPLC-MS 904

was performed on GBM samples from patients undergoing resection. Cortical tissue from an 905

epilepsy surgery was used as the control. (E) PDX GBM6 cells were growing in either 1% FBS 906

containing media or Neurobasal media (NBM). Conditioned media were collected at various 907

days and dopamine levels were calculated using HPLC. (F) PDX GBM43 cells were cultured in 908

the presence of either 20% or 1% oxygen (hypoxia) for three hours. Conditioned media was 909

collected, and dopamine levels determined. Dots represent values from each timepoint and/or 910

sample tested. Extended data figure 4-1 includes other genes involved in dopamine synthesis 911

(VMAT2, DAT, DDC, TH) measured in patient tumor samples as well as HPLC preformed on 912

various PDX subtypes undergoing a TMZ treatment time course. 913

914 915 Figure 5: DRD2 signaling activates hypoxia inducible factors in normoxic cultures. (A) 916

Correlation between DRD2 and 12042 genes from TCGA was determined by Pearson correlation 917

coefficients. 49 genes with coefficients >0.5 or <-0.5 and false discovery rate (FDR) <0.05 were 918

selected. Genes fitting these parameters were then analyzed using Enrichr. Top hits are shown in 919

the graph, with combined score and adjusted p value. (B) PDX GBM cells were treated with 920

DRD2 agonist (30nM) for 1, 2, or 4 days and then probed for expression of HIF1α by 921

immunoblot analysis. DMSO treated cells as vehicle treated control and cells exposed to hypoxic 922

conditions (1.5% O2) were used as a positive control for HIF expression, and beta actin was used 923

as a loading control. (C) Western blots were used to analyze the level of proteins in the VHL-924

PHD regulatory pathway after treatment with either DMSO or DRD2 agonist (20nM or 30nM). 925

Beta actin was used as a loading control. (D) PDX GBM6 were exposed to DRD2 agonist 926

Page 41: Activation of dopamine receptor 2 (DRD2) prompts

40

(30nm) and treated with HIF inhibitor for 24 48 and 72 hours SOX2, MMP2, and CMYC protein 927

levels were assayed. 928

929

Figure 6: DRD2 triggers alterations in gene expression and metabolic phenotype. (A) 930

Microarray analysis of gene expression was performed in PDX cells treated with DRD2 agonist 931

after four days. GeneVenn analysis was performed on gene transcripts found to be significantly 932

elevated in the two cell lines, revealing subtype specific alterations in expression profiles. (B) 933

Gene set enrichment analysis (GSEA) was performed on each cell line for canonical hypoxia 934

response signaling. (C/D) Microarray results were then limited to genes with confirmed hypoxia 935

response elements. (E/F) Seahorse analysis was performed to quantify the glycolytic rate in two 936

PDX lines, (E) GBM39, which responds to DRD2 activation with increased sphere-formation, 937

and (F) GBM12, which are unresponsive to DRD2 activation in neurosphere formation. Cells 938

were treated with either DMSO or 30nM DRD2 agonist and subjected to Seahorse analysis after 939

96 hours. Extended data figure 6-1 includes a responding PDX line, GBM5, as an assay control 940

demonstrating similar effects. Figure 6-1 also includes FACS analysis of glucose uptake and 941

fatty acid uptake in GBM12 and 39 as further validation of seahorse assays. (G) Summary 942

schematic of dopamine’s role within the tumor microenvironment and at the single cell level. 943

Microarray was performed on biological triplicates. Bars represent means from three 944

independent experiments and error bars represent SD. Student T-tests were performed for each 945

separate cell line. *P<.05, **P<.01, ***P<.001. 946

947

948

949

950

Page 42: Activation of dopamine receptor 2 (DRD2) prompts

41

951

Extended Data Figure 1-1: ChIP analysis of H3K27ac and H3K27me for other monoamine 952

receptors. 953

954

Extended Data Figure 1-2: GBM tumors express DRDs. (A) TCGA data reveals elevated 955

expression of DRD2 relative to the other DRDs. Dots represent mRNA levels from individual 956

tumors. Mean levels were compared using one-way ANOVA. (B) DRD2 expression in patient 957

samples varies based on subtype. (C) Expression of DRDs in PDX GBM cells was determined 958

using FACS analysis. Bars represent the mean of three individual experiments. (D) PDX GBM 959

cells were implanted intracranially into the right hemisphere of athymic nude mice. Dots 960

represent the mean from each individual mouse. 961

962

Extended Data Figure 3-1: Activation of DRD2 increases expression of GIC markers. (A) 963

Neurosphere assay of PDX GBM12 treated with either DRD2 against (30nM) or equimolar 964

DMSO. Extreme limiting dilution assays reveal no significant difference in stem cell frequency. 965

(B)Mice injected with DRD2+ cells show efficient tumor engraftment compared to DRD2- cells. 966

(C) PDX GBM cells treated with agonist or DMSO and Ki67 levels were determined by FACS. 967

No difference in Ki67 was detected. (D) PDX cells treated with agonist or DMSO and the 968

expression of GIC markers was examined by western blot after four days. Beta-actin was used as 969

a loading control. (E) U251-CD133 RFP driven reporter line was treated with agonist for 24 970

hours and RFP (reporter) and CD133 (ligand) were assayed. 971

972

Page 43: Activation of dopamine receptor 2 (DRD2) prompts

42

Extended Data Figure 4-1: (A) Patient tumors express mRNA for several genes involved in 973

synthesis, secretion, and reuptake of dopamine. Data were taken from the Cancer Genome Atlas 974

(TCGA). Expression was examined in astrocytomas (Grade 1, II, III, IV) and within GBM based 975

on subtype (Classical, Mesenchymal, Proneural). (B) PDX lines were exposed to 50μm TMZ 976

and collected after 2, 4, 6, and 8 days and subjected to HPLC. Results are shown normalized to 977

equimolar DMSO controls. 978

979

Extended Data Figure 6-1 (A) Alkylating chemotherapy alters the uptake of glucose and fatty 980

acid. PDX GBM cells were treated with either DMSO or TMZ. Four days later, cells were 981

exposed to fluorescently-labeled glucose and palmitate. Uptake was determined by FACS 982

analysis. (B) Seahorse analysis was used to determine the glycolytic rate of PDX lines after 4 983

days of 30nm agonist, or equimolar DMSO treatment. Comparisons were made using student t-984

Tests. *p<.05, **p<.01, ***p<.001. 985

Page 44: Activation of dopamine receptor 2 (DRD2) prompts
Page 45: Activation of dopamine receptor 2 (DRD2) prompts
Page 46: Activation of dopamine receptor 2 (DRD2) prompts
Page 47: Activation of dopamine receptor 2 (DRD2) prompts
Page 48: Activation of dopamine receptor 2 (DRD2) prompts
Page 49: Activation of dopamine receptor 2 (DRD2) prompts