In vivo brain GPCR signaling elucidated by phosphoproteomics

Mechanisms of drug action Advanced mass spectrometry methods enable monitoring of tens of thousands of phosphorylation sites in proteins. This technology can potentially distinguish cellular signaling pathways that produce beneficial effects from those that produce unwanted side effects. Liu et al. treated mice with various agonists of the kappa opioid receptor (a G protein–coupled receptor) and monitored changes in phosphorylation over time in different brain regions. The phosphorylation patterns revealed distinct patterns of signaling in various brain tissues, some of which were associated with unwanted side effects. Science, this issue p. eaao4927 High-throughput monitoring of phosphorylation helps define drug actions in the brain. INTRODUCTION The G protein–coupled receptor (GPCR) superfamily is a major drug target for neurological diseases. Functionally selective agonists activate GPCRs, such as the kappa opioid receptor (KOR), in a pathway-specific manner that may lead to drugs with fewer side effects. For example, KOR agonists that trigger beneficial antinociceptive, antipruritic, and anticonvulsant effects while causing minimal or no undesirable dysphoric, aversive, or psychotomimetic effects would be invaluable in light of the current opioid epidemic. However, functional selectivity observed in vitro frequently has little predictive value for behavioral outcomes. RATIONALE Obtaining a systems view of GPCR signaling in the brain may overcome the gap between in vitro pharmacology and in vivo testing. Recent breakthroughs in mass spectrometry–based proteomics have enabled us to quantify tens of thousands of phosphorylation events simultaneously in a high-throughput fashion. Using the KOR as a GPCR model, we applied this technology to achieve a global overview of the architecture of brain phosphoproteome in five mouse brain regions, in which we examined signaling induced by structurally and behaviorally diverse agonists. RESULTS Through the quantification of 50,000 different phosphosites, this approach yielded a brain region–specific systems view of the phosphoproteome, providing a context to understand KOR signaling in vivo. We observed strong regional specificity of KOR signaling attributable to differences in protein-protein interaction networks, neuronal contacts, and the different tissues in neuronal circuitries. Agonists with distinct signaling profiles elicited differential dynamic phosphorylation of synaptic proteins, thereby linking GPCR signaling to the modulation of brain functions. The most prominent changes occurred on synaptic proteins associated with dopaminergic, glutamatergic, and γ-aminobutyric acid–mediated (GABAergic) signaling and synaptic vesicle release. The large-scale dephosphorylation of synaptic proteins in the striatum after 5 min of agonist stimulation was partially blocked by protein phosphatase 2A (PP2A) inhibitors, underscoring the involvement of PP2A in KOR-mediated synaptic functions. Pathway analysis revealed enrichment of mTOR (mechanistic target of rapamycin) signaling by agonists associated with aversion. Strikingly, mTOR inhibition during KOR activation abolished aversion while preserving therapeutic antinociceptive and anticonvulsant effects. Parallel characterization of phosphoproteomic changes related to KOR-mediated mTOR activation in a cell line model provided additional mechanistic insights at the level of the signaling cascade. CONCLUSION We dissected the signaling pathways associated with desired and undesired outcomes of KOR activation in vivo and applied this knowledge to suppress the latter. Our work demonstrates the utility of combining phosphoproteomics with pharmacological tools and behavioral assessments as a general approach for studying GPCR signaling in vivo. Together with appropriate in vitro cellular systems, individual pathways can be characterized in depth, providing a rational basis for GPCR drug discovery. High-throughput phosphoproteomics to characterize in vivo brain GPCR signaling. Subsequent bioinformatic analysis enables prediction and modulation of downstream signaling pathways, which are correlated with unwanted effects but not the therapeutic outcome. A systems view of G protein–coupled receptor (GPCR) signaling in its native environment is central to the development of GPCR therapeutics with fewer side effects. Using the kappa opioid receptor (KOR) as a model, we employed high-throughput phosphoproteomics to investigate signaling induced by structurally diverse agonists in five mouse brain regions. Quantification of 50,000 different phosphosites provided a systems view of KOR in vivo signaling, revealing novel mechanisms of drug action. Thus, we discovered enrichment of the mechanistic target of rapamycin (mTOR) pathway by U-50,488H, an agonist causing aversion, which is a typical KOR-mediated side effect. Consequently, mTOR inhibition during KOR activation abolished aversion while preserving beneficial antinociceptive and anticonvulsant effects. Our results establish high-throughput phosphoproteomics as a general strategy to investigate GPCR in vivo signaling, enabling prediction and modulation of behavioral outcomes.

[1]  J. Kamei,et al.  Antipruritic activity of the κ-opioid receptor agonist, TRK-820 , 2002 .

[2]  Michael L. Creech,et al.  Integration of biological networks and gene expression data using Cytoscape , 2007, Nature Protocols.

[3]  Bryan L Roth,et al.  Structure-based design, synthesis, and biochemical and pharmacological characterization of novel salvinorin A analogues as active state probes of the kappa-opioid receptor. , 2009, Biochemistry.

[4]  C. Schwarzer 30 years of dynorphins--new insights on their functions in neuropsychiatric diseases. , 2009, Pharmacology & therapeutics.

[5]  A. Lymperopoulos,et al.  GPCR signaling and cardiac function. , 2015, European journal of pharmacology.

[6]  S. Gygi,et al.  Global phosphorylation analysis of β-arrestin–mediated signaling downstream of a seven transmembrane receptor (7TMR) , 2010, Proceedings of the National Academy of Sciences.

[7]  Michael L. Eads,et al.  Synergistic interactions of endogenous opioids and cannabinoid systems , 1999, Brain Research.

[8]  J. Kamei,et al.  Antipruritic activity of the kappa-opioid receptor agonist, TRK-820. , 2002, European journal of pharmacology.

[9]  L. Bohn,et al.  Investigation of the role of βarrestin2 in kappa opioid receptor modulation in a mouse model of pruritus , 2015, Neuropharmacology.

[10]  L. Bohn,et al.  Functional Selectivity of 6′-Guanidinonaltrindole (6′-GNTI) at κ-Opioid Receptors in Striatal Neurons* , 2013, The Journal of Biological Chemistry.

[11]  Chao Zhang,et al.  CB1 receptor antagonism prevents long-term hyperexcitability after head injury by regulation of dynorphin-KOR system and mGluR5 in rat hippocampus , 2016, Brain Research.

[12]  Ryan T. Strachan,et al.  Distinct Phosphorylation Sites on the β2-Adrenergic Receptor Establish a Barcode That Encodes Differential Functions of β-Arrestin , 2011, Science Signaling.

[13]  Henry Lin,et al.  Structure-based discovery of opioid analgesics with reduced side effects , 2016, Nature.

[14]  P. Taghert,et al.  Exploring the Biology of G Protein–Coupled Receptors from In Vitro to In Vivo , 2015, Molecular Pharmacology.

[15]  Angus C Nairn,et al.  Nicotine Regulates DARPP-32 (Dopamine- and cAMP-Regulated Phosphoprotein of 32 kDa) Phosphorylation at Multiple Sites in Neostriatal Neurons , 2005, Journal of Pharmacology and Experimental Therapeutics.

[16]  K. Inoki,et al.  TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling , 2002, Nature Cell Biology.

[17]  Ronald F. Mucha,et al.  Motivational properties of kappa and mu opioid receptor agonists studied with place and taste preference conditioning , 2004, Psychopharmacology.

[18]  M. Bruchas,et al.  Stress-Induced p38 Mitogen-Activated Protein Kinase Activation Mediates κ-Opioid-Dependent Dysphoria , 2007, The Journal of Neuroscience.

[19]  G. Sperk,et al.  Neuropeptide Y and somatostatin immunoreactivity in the rat hippocampus after moderate hypoxia , 1996, Naunyn-Schmiedeberg's Archives of Pharmacology.

[20]  M. Spetea,et al.  Highly Potent and Selective New Diphenethylamines Interacting with the κ-Opioid Receptor: Synthesis, Pharmacology, and Structure–Activity Relationships , 2017, Journal of medicinal chemistry.

[21]  S. Eans,et al.  Selective κ receptor partial agonist HS666 produces potent antinociception without inducing aversion after i.c.v. administration in mice , 2017, British journal of pharmacology.

[22]  Michael W. Salter,et al.  Src kinases: a hub for NMDA receptor regulation , 2004, Nature Reviews Neuroscience.

[23]  Mohammad Sayyah,et al.  Endogenous dynorphin in epileptogenesis and epilepsy: anticonvulsant net effect via kappa opioid receptors. , 2007, Brain : a journal of neurology.

[24]  M. Kuśmider,et al.  Potential role of G protein-coupled receptor (GPCR) heterodimerization in neuropsychiatric disorders: a focus on depression. , 2013, Pharmacological reports : PR.

[25]  W. Colmers,et al.  The G‐protein biased partial κ opioid receptor agonist 6′‐GNTI blocks hippocampal paroxysmal discharges without inducing aversion , 2016, British journal of pharmacology.

[26]  José A. Dianes,et al.  2016 update of the PRIDE database and its related tools , 2015, Nucleic Acids Res..

[27]  T. Shippenberg,et al.  Evidence that the aversive effects of opioid antagonists and κ-agonists are centrally mediated , 2004, Psychopharmacology.

[28]  Marco Y. Hein,et al.  The Perseus computational platform for comprehensive analysis of (prote)omics data , 2016, Nature Methods.

[29]  J. David Sweatt,et al.  Roles of serine/threonine phosphatases in hippocampel synaptic plasticity , 2001, Nature Reviews Neuroscience.

[30]  J. Coon,et al.  Phosphoproteomics in the Age of Rapid and Deep Proteome Profiling , 2015, Analytical chemistry.

[31]  Jamie Fong,et al.  A heterodimer-selective agonist shows in vivo relevance of G protein-coupled receptor dimers. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[32]  Ruedi Aebersold,et al.  Mass-spectrometric exploration of proteome structure and function , 2016, Nature.

[33]  D. Bashford,et al.  Liu F-NMR 19 by-Adrenergic Receptor Characterized 2 β Biased Signaling Pathways in , 2012 .

[34]  Tao Wang,et al.  SynaptomeDB: an ontology-based knowledgebase for synaptic genes , 2012, Bioinform..

[35]  A. Saria,et al.  Conditioned place preference for social interaction in rats: contribution of sensory components , 2011, Front. Behav. Neurosci..

[36]  J. Javitch,et al.  6′-Guanidinonaltrindole (6′-GNTI) Is a G Protein-biased κ-Opioid Receptor Agonist That Inhibits Arrestin Recruitment* , 2012, The Journal of Biological Chemistry.

[37]  Fahad Saeed,et al.  Dynamics of the G Protein-coupled Vasopressin V2 Receptor Signaling Network Revealed by Quantitative Phosphoproteomics* , 2011, Molecular & Cellular Proteomics.

[38]  M. Bruchas,et al.  Optogenetic approaches for dissecting neuromodulation and GPCR signaling in neural circuits. , 2017, Current opinion in pharmacology.

[39]  L. Bohn,et al.  β-arrestins: regulatory role and therapeutic potential in opioid and cannabinoid receptor-mediated analgesia. , 2014, Handbook of experimental pharmacology.

[40]  C. Chen,et al.  Cloning of a human kappa opioid receptor from the brain. , 1995, Life sciences.

[41]  Kurt Wüthrich,et al.  Biased Signaling Pathways in β2-Adrenergic Receptor Characterized by 19F-NMR , 2012, Science.

[42]  M. Mann,et al.  Ultradeep human phosphoproteome reveals a distinct regulatory nature of Tyr and Ser/Thr-based signaling. , 2014, Cell reports.

[43]  Matthias Mann,et al.  Cell type– and brain region–resolved mouse brain proteome , 2015, Nature Neuroscience.

[44]  Bin Zhang,et al.  PhosphoSitePlus: a comprehensive resource for investigating the structure and function of experimentally determined post-translational modifications in man and mouse , 2011, Nucleic Acids Res..

[45]  P. Yadav,et al.  Biased agonism at kappa opioid receptors: Implication in pain and mood disorders. , 2015, European journal of pharmacology.

[46]  B. Roth,et al.  The G Protein–Biased κ-Opioid Receptor Agonist RB-64 Is Analgesic with a Unique Spectrum of Activities In Vivo , 2015, The Journal of Pharmacology and Experimental Therapeutics.

[47]  José A. Dianes,et al.  2016 update of the PRIDE database and its related tools , 2016, Nucleic Acids Res..

[48]  M. Mann,et al.  Proteomic maps of breast cancer subtypes , 2016, Nature Communications.

[49]  Jüergen Cox,et al.  The MaxQuant computational platform for mass spectrometry-based shotgun proteomics , 2016, Nature Protocols.

[50]  D. Ferguson,et al.  Transformation of a κ-Opioid Receptor Antagonist to a κ-Agonist by Transfer of a Guanidinium Group from the 5‘- to 6‘-Position of Naltrindole , 2001 .

[51]  M. Ashburner,et al.  Gene Ontology: tool for the unification of biology , 2000, Nature Genetics.

[52]  M. Austin,et al.  The mTOR signaling pathway in the prefrontal cortex is compromised in major depressive disorder , 2011, Progress in Neuro-Psychopharmacology and Biological Psychiatry.

[53]  Paul Greengard,et al.  A phosphatase cascade by which rewarding stimuli control nucleosomal response , 2008, Nature.

[54]  Hey-Kyoung Lee,et al.  Synaptic plasticity and phosphorylation. , 2006, Pharmacology & therapeutics.

[55]  M. Spetea,et al.  Discovery and biological evaluation of a diphenethylamine derivative (HS665), a highly potent and selective κ opioid receptor agonist , 2012, BMC Pharmacology and Toxicology.

[56]  Everolimus improves neuropsychiatric symptoms in a patient with tuberous sclerosis carrying a novel TSC2 mutation , 2016, Molecular Brain.

[57]  T. Shippenberg,et al.  Evidence that the aversive effects of opioid antagonists and kappa-agonists are centrally mediated. , 1989, Psychopharmacology.

[58]  Sean J. Humphrey,et al.  Phosphorylation Is a Central Mechanism for Circadian Control of Metabolism and Physiology. , 2017, Cell metabolism.

[59]  Chongguang Chen,et al.  Cloning of a human κ opioid receptor from the brain , 1995 .

[60]  M. Janech,et al.  The β-Arrestin Pathway-selective Type 1A Angiotensin Receptor (AT1A) Agonist [Sar1,Ile4,Ile8]Angiotensin II Regulates a Robust G Protein-independent Signaling Network* , 2011, The Journal of Biological Chemistry.

[61]  R. M. Jones,et al.  Transformation of a kappa-opioid receptor antagonist to a kappa-agonist by transfer of a guanidinium group from the 5'- to 6'-position of naltrindole. , 2001, Journal of medicinal chemistry.

[62]  L. Mansfield,et al.  A focus on depression , 2019 .

[63]  Sara R. Jones,et al.  Biased agonists of the kappa opioid receptor suppress pain and itch without causing sedation or dysphoria , 2016, Science Signaling.

[64]  R. Lahti,et al.  U-50,488: a selective and structurally novel non-Mu (kappa) opioid agonist. , 1983, The Journal of pharmacology and experimental therapeutics.

[65]  B. Roth,et al.  Identification of Novel Functionally Selective κ-Opioid Receptor Scaffolds , 2014, Molecular Pharmacology.

[66]  Virginia Chu,et al.  Towards the miniaturization of GPCR-based live-cell screening assays. , 2012, Trends in biotechnology.

[67]  Allison T. Knoll,et al.  Dynorphin, stress, and depression , 2010, Brain Research.

[68]  J. Gutkind,et al.  G-protein-coupled receptors and cancer , 2007, Nature Reviews Cancer.

[69]  Marco Y. Hein,et al.  Accurate Proteome-wide Label-free Quantification by Delayed Normalization and Maximal Peptide Ratio Extraction, Termed MaxLFQ * , 2014, Molecular & Cellular Proteomics.

[70]  A. Heck,et al.  Next-generation proteomics: towards an integrative view of proteome dynamics , 2012, Nature Reviews Genetics.

[71]  P. Greengard,et al.  Dopamine and cAMP-Regulated Phosphoprotein 32 kDa Controls Both Striatal Long-Term Depression and Long-Term Potentiation, Opposing Forms of Synaptic Plasticity , 2000, The Journal of Neuroscience.

[72]  P. Phillips,et al.  Kappa Opioid Receptor-Induced Aversion Requires p38 MAPK Activation in VTA Dopamine Neurons , 2015, The Journal of Neuroscience.

[73]  R. Hampson,et al.  Cannabinoid and kappa opioid receptors reduce potassium K current via activation of G(s) proteins in cultured hippocampal neurons. , 2000, Journal of neurophysiology.

[74]  R. Burgoyne,et al.  Protein phosphorylation and the regulation of synaptic membrane traffic , 1999, Trends in Neurosciences.

[75]  B. Roques,et al.  Disruption of the κ‐opioid receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective κ‐agonist U‐50,488H and attenuates morphine withdrawal , 1998, The EMBO journal.

[76]  S. Snyder,et al.  Antidepressant action of ketamine via mTOR is mediated by inhibition of nitrergic Rheb degradation , 2016, Molecular Psychiatry.

[77]  G. Milligan,et al.  Deconvolution of complex G protein–coupled receptor signaling in live cells using dynamic mass redistribution measurements , 2010, Nature Biotechnology.

[78]  J. Lipton,et al.  The Neurology of mTOR , 2014, Neuron.

[79]  S. Gammeltoft,et al.  Quantitative Phosphoproteomics Dissection of Seven-transmembrane Receptor Signaling Using Full and Biased Agonists* , 2010, Molecular & Cellular Proteomics.

[80]  Sean J Humphrey,et al.  High-throughput phosphoproteomics reveals in vivo insulin signaling dynamics , 2015, Nature Biotechnology.