Inhibition of phosphodiesterase 4D suppresses mTORC1 signaling and pancreatic cancer growth

The mammalian target of rapamycin complex 1 (mTORC1) senses multiple upstream stimuli to orchestrate anabolic and catabolic events that regulate cell growth and metabolism. Hyperactivation of mTORC1 signaling is observed in multiple human diseases; thus, pathways that suppress mTORC1 signaling may help to identify new therapeutic targets. Here, we report that phosphodiesterase 4D (PDE4D) promotes pancreatic cancer tumor growth by increasing mTORC1 signaling. GPCRs paired to Gαs proteins activate adenylyl cyclase, which in turn elevates levels of 3′,5′-cyclic adenosine monophosphate (cAMP), whereas PDEs catalyze the hydrolysis of cAMP to 5′-AMP. PDE4D forms a complex with mTORC1 and is required for mTORC1 lysosomal localization and activation. Inhibition of PDE4D and the elevation of cAMP levels block mTORC1 signaling via Raptor phosphorylation. Moreover, pancreatic cancer exhibits an upregulation of PDE4D expression, and high PDE4D levels predict the poor overall survival of patients with pancreatic cancer. Importantly, FDA-approved PDE4 inhibitors repress pancreatic cancer cell tumor growth in vivo by suppressing mTORC1 signaling. Our results identify PDE4D as an important activator of mTORC1 and suggest that targeting PDE4 with FDA-approved inhibitors may be beneficial for the treatment of human diseases with hyperactivated mTORC1 signaling.

[1]  Shihai Zhang,et al.  AKAP13 couples GPCR signaling to mTORC1 inhibition , 2021, PLoS genetics.

[2]  H. Hua,et al.  Complex roles of cAMP–PKA–CREB signaling in cancer , 2020, Experimental Hematology & Oncology.

[3]  Jenna L. Jewell,et al.  Regulation of mTORC1 by Upstream Stimuli , 2020, Genes.

[4]  M. Maki,et al.  Amino acid-dependent control of mTORC1 signaling: a variety of regulatory modes , 2020, Journal of Biomedical Science.

[5]  T. van Groen,et al.  Dominant-Negative Attenuation of cAMP-Selective Phosphodiesterase PDE4D Action Affects Learning and Behavior , 2020, International journal of molecular sciences.

[6]  M. Gurney,et al.  PDE4 subtypes in cancer , 2020, Oncogene.

[7]  J. Phillips Inhaled Phosphodiesterase 4 (PDE4) Inhibitors for Inflammatory Respiratory Diseases , 2020, Frontiers in Pharmacology.

[8]  Delong Meng,et al.  Glutamine and asparagine activate mTORC1 independently of Rag GTPases , 2020, The Journal of Biological Chemistry.

[9]  Zhi Li,et al.  High expression of PDE4D correlates with poor prognosis and clinical progression in pancreaticductal adenocarcinoma , 2019, Journal of Cancer.

[10]  H. Hua,et al.  Targeting mTOR for cancer therapy , 2019, Journal of Hematology & Oncology.

[11]  B. Manning,et al.  Molecular logic of mTORC1 signalling as a metabolic rheostat , 2019, Nature Metabolism.

[12]  Jinhua Zhang,et al.  mTOR Signaling in Cancer and mTOR Inhibitors in Solid Tumor Targeting Therapy , 2019, International journal of molecular sciences.

[13]  Sang-Woo Kim,et al.  Phosphodiesterase 4B is an effective therapeutic target in colorectal cancer. , 2019, Biochemical and biophysical research communications.

[14]  K. Guan,et al.  mTOR as a central hub of nutrient signalling and cell growth , 2019, Nature Cell Biology.

[15]  Susan S. Taylor,et al.  GPCR signaling inhibits mTORC1 via PKA phosphorylation of Raptor , 2019, eLife.

[16]  Heng Li,et al.  Phosphodiesterase-4 Inhibitors for the Treatment of Inflammatory Diseases , 2018, Front. Pharmacol..

[17]  D. Sabatini,et al.  Ragulator and SLC38A9 activate the Rag GTPases through noncanonical GEF mechanisms , 2018, Proceedings of the National Academy of Sciences.

[18]  W. Weis,et al.  The Molecular Basis of G Protein-Coupled Receptor Activation. , 2018, Annual review of biochemistry.

[19]  Xin Wei,et al.  Inhibitors of phosphodiesterase as cancer therapeutics. , 2018, European journal of medicinal chemistry.

[20]  Paul A. Insel,et al.  G Protein-Coupled Receptors as Targets for Approved Drugs: How Many Targets and How Many Drugs? , 2018, Molecular Pharmacology.

[21]  E. Klussmann,et al.  Roles of A-Kinase Anchoring Proteins and Phosphodiesterases in the Cardiovascular System , 2018, Journal of cardiovascular development and disease.

[22]  Ö. Sahin,et al.  Reactivation of cAMP Pathway by PDE4D Inhibition Represents a Novel Druggable Axis for Overcoming Tamoxifen Resistance in ER-positive Breast Cancer , 2018, Clinical Cancer Research.

[23]  G. Baillie,et al.  PDE4-Mediated cAMP Signalling , 2018, Journal of cardiovascular development and disease.

[24]  N. Pavletich,et al.  Structural Mechanisms of mTORC1 activation by RHEB and inhibition by PRAS40 , 2017, Nature.

[25]  N. Perrimon,et al.  mTORC1 Couples Nucleotide Synthesis to Nucleotide Demand Resulting in a Targetable Metabolic Vulnerability. , 2017, Cancer cell.

[26]  Chin-Rang Yang,et al.  Systems-level identification of PKA-dependent signaling in epithelial cells , 2017, Proceedings of the National Academy of Sciences.

[27]  Jenna L. Jewell,et al.  Amino acid and small GTPase regulation of mTORC1 , 2017, Cellular logistics.

[28]  Line Victoria Moen,et al.  Evolution of the cAMP-dependent protein kinase (PKA) catalytic subunit isoforms , 2017, PloS one.

[29]  Cheng Li,et al.  GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses , 2017, Nucleic Acids Res..

[30]  David M. Sabatini,et al.  mTOR Signaling in Growth, Metabolism, and Disease , 2017, Cell.

[31]  Gregory A. Wyant,et al.  KICSTOR recruits GATOR1 to the lysosome and is necessary for nutrients to regulate mTORC1 , 2017, Nature.

[32]  O. Brady,et al.  Rags to riches: Amino acid sensing by the Rag GTPases in health and disease , 2016, Small GTPases.

[33]  M. Conti,et al.  PDE4D phosphorylation: A coincidence detector integrating multiple signaling pathways. , 2016, Cellular signalling.

[34]  E. Kennedy,et al.  Defining A‐Kinase Anchoring Protein (AKAP) Specificity for the Protein Kinase A Subunit RI (PKA‐RI) , 2016, Chembiochem : a European journal of chemical biology.

[35]  Rohit Rao,et al.  The G protein Gαs acts as a tumor suppressor in sonic hedgehog signaling-driven tumorigenesis , 2016, Cell cycle.

[36]  Jia-You Fang,et al.  Recent Advances Using Phosphodiesterase 4 (PDE4) Inhibitors to Treat Inflammatory Disorders: Animal and Clinical Studies , 2016 .

[37]  Yuan-Lu Cui,et al.  The cyclic AMP signaling pathway: Exploring targets for successful drug discovery (Review) , 2016, Molecular medicine reports.

[38]  J. Blenis,et al.  A nexus for cellular homeostasis: the interplay between metabolic and signal transduction pathways. , 2015, Current opinion in biotechnology.

[39]  A. Karginov,et al.  UCR1C is a novel activator of phosphodiesterase 4 (PDE4) long isoforms and attenuates cardiomyocyte hypertrophy. , 2015, Cellular signalling.

[40]  Susan S. Taylor,et al.  Inactivation of a Gαs-PKA tumor suppressor pathway in skin stem cells initiates basal-cell carcinogenesis , 2015, Nature Cell Biology.

[41]  M. Vieytes,et al.  Key role of phosphodiesterase 4A (PDE4A) in autophagy triggered by yessotoxin. , 2015, Toxicology.

[42]  K. Guan,et al.  Differential regulation of mTORC1 by leucine and glutamine , 2015, Science.

[43]  Michael N. Hall,et al.  mTORC1 mediated translational elongation limits intestinal tumour initiation and growth , 2014, Nature.

[44]  D. Beebe,et al.  Phosphodiesterase 4D Inhibitors Limit Prostate Cancer Growth Potential , 2014, Molecular Cancer Research.

[45]  L. Sapio,et al.  Targeting protein kinase A in cancer therapy: an update , 2014, EXCLI journal.

[46]  M. Kool,et al.  The G-protein Alpha Subunit Gsα Is A Tumor Suppressor In Sonic Hedgehog-driven Medulloblastoma , 2014, Nature Medicine.

[47]  D. Sabatini,et al.  Regulation of mTORC1 by amino acids. , 2014, Trends in cell biology.

[48]  Aurélien Rizk,et al.  Segmentation and quantification of subcellular structures in fluorescence microscopy images using Squassh , 2014, Nature Protocols.

[49]  C. Betz,et al.  Where is mTOR and what is it doing there? , 2013, The Journal of cell biology.

[50]  D. Sabatini,et al.  The folliculin tumor suppressor is a GAP for the RagC/D GTPases that signal amino acid levels to mTORC1. , 2013, Molecular cell.

[51]  F. Zwartkruis,et al.  Rheb and Rags come together at the lysosome to activate mTORC1. , 2013, Biochemical Society transactions.

[52]  D. Sabatini,et al.  Regulation of mTORC1 and its impact on gene expression at a glance , 2013, Journal of Cell Science.

[53]  Matthew Meyerson,et al.  A Tumor Suppressor Complex with GAP Activity for the Rag GTPases That Signal Amino Acid Sufficiency to mTORC1 , 2013, Science.

[54]  W. Seeger,et al.  Phosphodiesterase-4 promotes proliferation and angiogenesis of lung cancer by crosstalk with HIF , 2013, Oncogene.

[55]  K. Guan,et al.  Amino acid signalling upstream of mTOR , 2013, Nature Reviews Molecular Cell Biology.

[56]  Paolo Sassone-Corsi,et al.  The cyclic AMP pathway. , 2012, Cold Spring Harbor perspectives in biology.

[57]  D. Sabatini,et al.  Ragulator Is a GEF for the Rag GTPases that Signal Amino Acid Levels to mTORC1 , 2012, Cell.

[58]  Vassilios Ioannidis,et al.  ExPASy: SIB bioinformatics resource portal , 2012, Nucleic Acids Res..

[59]  C. Lugnier,et al.  Cyclic nucleotide phosphodiesterase (PDE) isozymes as targets of the intracellular signalling network: benefits of PDE inhibitors in various diseases and perspectives for future therapeutic developments , 2012, British journal of pharmacology.

[60]  H. Ke,et al.  Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases , 2012, Cell.

[61]  E. Fedele,et al.  GEBR‐7b, a novel PDE4D selective inhibitor that improves memory in rodents at non‐emetic doses , 2011, British journal of pharmacology.

[62]  Roberto Zoncu,et al.  mTORC1 Senses Lysosomal Amino Acids Through an Inside-Out Mechanism That Requires the Vacuolar H+-ATPase , 2011, Science.

[63]  John D. Scott,et al.  Discovery of cellular substrates for protein kinase A using a peptide array screening protocol. , 2011, The Biochemical journal.

[64]  K. Rabe Update on roflumilast, a phosphodiesterase 4 inhibitor for the treatment of chronic obstructive pulmonary disease , 2011, British journal of pharmacology.

[65]  M. Conti,et al.  PDE4D and PDE4B Function in Distinct Subcellular Compartments in Mouse Embryonic Fibroblasts* , 2011, The Journal of Biological Chemistry.

[66]  D. Sabatini,et al.  mTOR: from growth signal integration to cancer, diabetes and ageing , 2010, Nature Reviews Molecular Cell Biology.

[67]  D. Sabatini,et al.  Ragulator-Rag Complex Targets mTORC1 to the Lysosomal Surface and Is Necessary for Its Activation by Amino Acids , 2010, Cell.

[68]  D. Sabatini,et al.  mTOR signaling at a glance , 2009, Journal of Cell Science.

[69]  F. Reimann,et al.  The role of the PDE4D cAMP phosphodiesterase in the regulation of glucagon‐like peptide‐1 release , 2009, British journal of pharmacology.

[70]  Ralph Weissleder,et al.  Effective Use of PI3K and MEK Inhibitors to Treat Mutant K-Ras G12D and PIK3CA H1047R Murine Lung Cancers , 2008, Nature Medicine.

[71]  David M. Sabatini,et al.  The Rag GTPases Bind Raptor and Mediate Amino Acid Signaling to mTORC1 , 2008, Science.

[72]  G. Baillie,et al.  cAMP-Specific phosphodiesterase-4 enzymes in the cardiovascular system: a molecular toolbox for generating compartmentalized cAMP signaling. , 2007, Circulation research.

[73]  Mike Tyers,et al.  BioGRID: a general repository for interaction datasets , 2005, Nucleic Acids Res..

[74]  S. Jin,et al.  Splice variants of the cyclic nucleotide phosphodiesterase PDE4D are differentially expressed and regulated in rat tissue. , 2005, The Biochemical journal.

[75]  Joseph Avruch,et al.  Rheb Binds and Regulates the mTOR Kinase , 2005, Current Biology.

[76]  J. Avruch,et al.  Dissociation of raptor from mTOR is a mechanism of rapamycin‐induced inhibition of mTOR function , 2004, Genes to cells : devoted to molecular & cellular mechanisms.

[77]  L. Cantley,et al.  Rheb fills a GAP between TSC and TOR. , 2003, Trends in biochemical sciences.

[78]  K. Inoki,et al.  Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. , 2003, Genes & development.

[79]  Paul Tempst,et al.  GbetaL, a positive regulator of the rapamycin-sensitive pathway required for the nutrient-sensitive interaction between raptor and mTOR. , 2003, Molecular cell.

[80]  G. Livera,et al.  Cyclic AMP-specific PDE4 Phosphodiesterases as Critical Components of Cyclic AMP Signaling* , 2003, The Journal of Biological Chemistry.

[81]  J. Avruch,et al.  Raptor, a Binding Partner of Target of Rapamycin (TOR), Mediates TOR Action , 2002, Cell.

[82]  Daniel McCormick,et al.  Phosphorylation of Centrin during the Cell Cycle and Its Role in Centriole Separation Preceding Centrosome Duplication* , 2001, The Journal of Biological Chemistry.

[83]  M. Houslay,et al.  Action of rolipram on specific PDE4 cAMP phosphodiesterase isoforms and on the phosphorylation of cAMP-response-element-binding protein (CREB) and p38 mitogen-activated protein (MAP) kinase in U937 monocytic cells. , 2000, The Biochemical journal.

[84]  M. Houslay,et al.  UCR1 and UCR2 Domains Unique to the cAMP-specific Phosphodiesterase Family Form a Discrete Module via Electrostatic Interactions* , 2000, The Journal of Biological Chemistry.

[85]  E B Cox,et al.  Use of a monoclonal anti-estrogen receptor antibody in the immunohistochemical evaluation of human tumors. , 1986, Cancer research.

[86]  B. Kobilka,et al.  Structure and dynamics of GPCR signaling complexes , 2017, Nature Structural & Molecular Biology.