1
|
Paes D, Schepers M, Willems E, Rombaut B, Tiane A, Solomina Y, Tibbo A, Blair C, Kyurkchieva E, Baillie GS, Ricciarelli R, Brullo C, Fedele E, Bruno O, van den Hove D, Vanmierlo T, Prickaerts J. Ablation of specific long PDE4D isoforms increases neurite elongation and conveys protection against amyloid-β pathology. Cell Mol Life Sci 2023; 80:178. [PMID: 37306762 DOI: 10.1007/s00018-023-04804-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 06/13/2023]
Abstract
Inhibition of phosphodiesterase 4D (PDE4D) enzymes has been investigated as therapeutic strategy to treat memory problems in Alzheimer's disease (AD). Although PDE4D inhibitors are effective in enhancing memory processes in rodents and humans, severe side effects may hamper their clinical use. PDE4D enzymes comprise different isoforms, which, when targeted specifically, can increase treatment efficacy and safety. The function of PDE4D isoforms in AD and in molecular memory processes per se has remained unresolved. Here, we report the upregulation of specific PDE4D isoforms in transgenic AD mice and hippocampal neurons exposed to amyloid-β. Furthermore, by means of pharmacological inhibition and CRISPR-Cas9 knockdown, we show that the long-form PDE4D3, -D5, -D7, and -D9 isoforms regulate neuronal plasticity and convey resilience against amyloid-β in vitro. These results indicate that isoform-specific, next to non-selective, PDE4D inhibition is efficient in promoting neuroplasticity in an AD context. Therapeutic effects of non-selective PDE4D inhibitors are likely achieved through actions on long isoforms. Future research should identify which long PDE4D isoforms should be specifically targeted in vivo to both improve treatment efficacy and reduce side effects.
Collapse
Affiliation(s)
- Dean Paes
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Melissa Schepers
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Emily Willems
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Ben Rombaut
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Assia Tiane
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Yevgeniya Solomina
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Amy Tibbo
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Connor Blair
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Elka Kyurkchieva
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - George S Baillie
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Roberta Ricciarelli
- Section of General Pathology, Department of Experimental Medicine, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Chiara Brullo
- Section of Medicinal Chemistry, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Ernesto Fedele
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Section of Pharmacology and Toxicology, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Olga Bruno
- Section of Medicinal Chemistry, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Daniel van den Hove
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Tim Vanmierlo
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.
| | - Jos Prickaerts
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
2
|
Paes D, Schepers M, Rombaut B, van den Hove D, Vanmierlo T, Prickaerts J. The Molecular Biology of Phosphodiesterase 4 Enzymes as Pharmacological Targets: An Interplay of Isoforms, Conformational States, and Inhibitors. Pharmacol Rev 2021; 73:1016-1049. [PMID: 34233947 DOI: 10.1124/pharmrev.120.000273] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The phosphodiesterase 4 (PDE4) enzyme family plays a pivotal role in regulating levels of the second messenger cAMP. Consequently, PDE4 inhibitors have been investigated as a therapeutic strategy to enhance cAMP signaling in a broad range of diseases, including several types of cancers, as well as in various neurologic, dermatological, and inflammatory diseases. Despite their widespread therapeutic potential, the progression of PDE4 inhibitors into the clinic has been hampered because of their related relatively small therapeutic window, which increases the chance of producing adverse side effects. Interestingly, the PDE4 enzyme family consists of several subtypes and isoforms that can be modified post-translationally or can engage in specific protein-protein interactions to yield a variety of conformational states. Inhibition of specific PDE4 subtypes, isoforms, or conformational states may lead to more precise effects and hence improve the safety profile of PDE4 inhibition. In this review, we provide an overview of the variety of PDE4 isoforms and how their activity and inhibition is influenced by post-translational modifications and interactions with partner proteins. Furthermore, we describe the importance of screening potential PDE4 inhibitors in view of different PDE4 subtypes, isoforms, and conformational states rather than testing compounds directed toward a specific PDE4 catalytic domain. Lastly, potential mechanisms underlying PDE4-mediated adverse effects are outlined. In this review, we illustrate that PDE4 inhibitors retain their therapeutic potential in myriad diseases, but target identification should be more precise to establish selective inhibition of disease-affected PDE4 isoforms while avoiding isoforms involved in adverse effects. SIGNIFICANCE STATEMENT: Although the PDE4 enzyme family is a therapeutic target in an extensive range of disorders, clinical use of PDE4 inhibitors has been hindered because of the adverse side effects. This review elaborately shows that safer and more effective PDE4 targeting is possible by characterizing 1) which PDE4 subtypes and isoforms exist, 2) how PDE4 isoforms can adopt specific conformations upon post-translational modifications and protein-protein interactions, and 3) which PDE4 inhibitors can selectively bind specific PDE4 subtypes, isoforms, and/or conformations.
Collapse
Affiliation(s)
- Dean Paes
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Melissa Schepers
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Ben Rombaut
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Daniel van den Hove
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Tim Vanmierlo
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Jos Prickaerts
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| |
Collapse
|
3
|
Zuo H, Cattani-Cavalieri I, Musheshe N, Nikolaev VO, Schmidt M. Phosphodiesterases as therapeutic targets for respiratory diseases. Pharmacol Ther 2019; 197:225-242. [PMID: 30759374 DOI: 10.1016/j.pharmthera.2019.02.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD) and asthma, affect millions of people all over the world. Cyclic adenosine monophosphate (cAMP) which is one of the most important second messengers, plays a vital role in relaxing airway smooth muscles and suppressing inflammation. Given its vast role in regulating intracellular responses, cAMP provides an attractive pharmaceutical target in the treatment of chronic respiratory diseases. Phosphodiesterases (PDEs) are enzymes that hydrolyze cyclic nucleotides and help control cyclic nucleotide signals in a compartmentalized manner. Currently, the selective PDE4 inhibitor, roflumilast, is used as an add-on treatment for patients with severe COPD associated with bronchitis and a history of frequent exacerbations. In addition, other novel PDE inhibitors are in different phases of clinical trials. The current review provides an overview of the regulation of various PDEs and the potential application of selective PDE inhibitors in the treatment of COPD and asthma. The possibility to combine various PDE inhibitors as a way to increase their therapeutic effectiveness is also emphasized.
Collapse
Affiliation(s)
- Haoxiao Zuo
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Institute of Experimental Cardiovascular Research, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Isabella Cattani-Cavalieri
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nshunge Musheshe
- Department of Molecular Pharmacology, University of Groningen, the Netherlands
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany; German Center for Cardiovascular Research (DZHK), 20246 Hamburg, Germany
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
4
|
Briet C, Pereda A, Le Stunff C, Motte E, de Dios Garcia-Diaz J, de Nanclares GP, Dumaz N, Silve C. Mutations causing acrodysostosis-2 facilitate activation of phosphodiesterase 4D3. Hum Mol Genet 2018; 26:3883-3894. [PMID: 29016851 DOI: 10.1093/hmg/ddx271] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 07/08/2017] [Indexed: 01/21/2023] Open
Abstract
Type 2 acrodysostosis (ACRDYS2), a rare developmental skeletal dysplasia characterized by short stature, severe brachydactyly and facial dysostosis, is caused by mutations in the phosphodiesterase (PDE) 4D (PDE4D) gene. Several arguments suggest that the mutations should result in inappropriately increased PDE4D activity, however, no direct evidence supporting this hypothesis has been presented, and the functional consequences of the mutations remain unclear. We evaluated the impact of four different PDE4D mutations causing ACRDYS2 located in different functional domains on the activity of PDE4D3 expressed in Chinese hamster ovary cells. Three independent approaches were used: the direct measurement of PDE activity in cell lysates, the evaluation of intracellular cAMP levels using an EPAC-based (exchange factor directly activated by cAMP) bioluminescence resonance energy transfer sensor , and the assessment of PDE4D3 activation based on electrophoretic mobility. Our findings indicate that PDE4D3s carrying the ACRDYS2 mutations are more easily activated by protein kinase A-induced phosphorylation than WT PDE4D3. This occurs over a wide range of intracellular cAMP concentrations, including basal conditions, and result in increased hydrolytic activity. Our results provide new information concerning the mechanism whereby the mutations identified in the ACRDYS2 dysregulate PDE4D activity, and give insights into rare diseases involving the cAMP signaling pathway. These findings may offer new perspectives into the selection of specific PDE inhibitors and possible therapeutic intervention for these patients.
Collapse
Affiliation(s)
- Claire Briet
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Endocrinology, Diabetology and Nutrition, Mitovasc Institute, CHU Angers, France
| | - Arrate Pereda
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, OSI Araba-Txagorritxu, E-01009 Vitoria-Gasteiz, Spain
| | - Catherine Le Stunff
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Emmanuelle Motte
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Université Versailles - St Quentin, UFR des Sciences de la santé Simone Veil, Versailles, Paris, France
| | - Juan de Dios Garcia-Diaz
- Clinical Genetics Unit, Department of Internal Medicine, University Hospital Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Guiomar Perez de Nanclares
- Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, OSI Araba-Txagorritxu, E-01009 Vitoria-Gasteiz, Spain
| | - Nicolas Dumaz
- INSERM U976, Institut de Recherche sur la Peau, Hôpital Saint Louis, Paris, France
| | - Caroline Silve
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Centre de Référence des Maladies Rares du Métabolisme du Phosphore et du Calcium/Filière de Santé Maladies Rares OSCAR, Assistance Publique Hôpitaux de Paris, Paris, France.,Service de Biochimie et Génétique Moléculaires, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France
| |
Collapse
|
5
|
Böttcher R, Dulla K, van Strijp D, Dits N, Verhoef EI, Baillie GS, van Leenders GJLH, Houslay MD, Jenster G, Hoffmann R. Human PDE4D isoform composition is deregulated in primary prostate cancer and indicative for disease progression and development of distant metastases. Oncotarget 2018; 7:70669-70684. [PMID: 27683107 PMCID: PMC5342582 DOI: 10.18632/oncotarget.12204] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 09/12/2016] [Indexed: 02/07/2023] Open
Abstract
Phosphodiesterase 4D7 was recently shown to be specifically over-expressed in localized prostate cancer, raising the question as to which regulatory mechanisms are involved and whether other isoforms of this gene family (PDE4D) are affected under the same conditions.We investigated PDE4D isoform composition in prostatic tissues using a total of seven independent expression datasets and also included data on DNA methylation, copy number and AR and ERG binding in PDE4D promoters to gain insight into their effect on PDE4D transcription.We show that expression of PDE4D isoforms is consistently altered in primary human prostate cancer compared to benign tissue, with PDE4D7 being up-regulated while PDE4D5 and PDE4D9 are down-regulated. Disease progression is marked by an overall down-regulation of long PDE4D isoforms, while short isoforms (PDE4D1/2) appear to be relatively unaffected. While these alterations seem to be independent of copy number alterations in the PDE4D locus and driven by AR and ERG binding, we also observed increased DNA methylation in the promoter region of PDE4D5, indicating a long lasting alteration of the isoform composition in prostate cancer tissues.We propose two independent metrics that may serve as diagnostic and prognostic markers for prostate disease: (PDE4D7 - PDE4D5) provides an effective means for distinguishing PCa from normal adjacent prostate, whereas PDE4D1/2 - (PDE4D5 + PDE4D7 + PDE4D9) offers strong prognostic potential to detect aggressive forms of PCa and is associated with metastasis free survival. Overall, our findings highlight the relevance of PDE4D as prostate cancer biomarker and potential drug target.
Collapse
Affiliation(s)
- René Böttcher
- Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Bioinformatics, Technical University of Applied Sciences Wildau, Wildau, Germany
| | - Kalyan Dulla
- Department of Oncology Solutions and Precision Diagnostics, Philips Research Europe, Eindhoven, The Netherlands
| | - Dianne van Strijp
- Department of Oncology Solutions and Precision Diagnostics, Philips Research Europe, Eindhoven, The Netherlands
| | - Natasja Dits
- Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Esther I Verhoef
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - George S Baillie
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, Scotland, UK
| | | | - Miles D Houslay
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Guido Jenster
- Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ralf Hoffmann
- Department of Oncology Solutions and Precision Diagnostics, Philips Research Europe, Eindhoven, The Netherlands.,Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, Scotland, UK
| |
Collapse
|
6
|
Hoppmann J, Gesing J, Silve C, Leroy C, Bertsche A, Hirsch FW, Kiess W, Pfäffle R, Schuster V. Phenotypic Variability in a Family with Acrodysostosis Type 2 Caused by a Novel PDE4D Mutation Affecting the Serine Target of Protein Kinase-A Phosphorylation. J Clin Res Pediatr Endocrinol 2017; 9:360-365. [PMID: 28515031 PMCID: PMC5785644 DOI: 10.4274/jcrpe.4488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Acrodysostosis is a very rare congenital multisystem condition characterized by skeletal dysplasia with severe brachydactyly, midfacial hypoplasia, and short stature, varying degrees of intellectual disability, and possible resistance to multiple G protein-coupled receptor signalling hormones. Two distinct subtypes are differentiated: acrodysostosis type 1 resulting from defects in protein kinase type 1-α regulatory subunit and acrodysostosis type 2 caused by mutations in phosphodiesterase 4D (PDE4D). Most cases are sporadic. We report on a rare multigenerational familial case of acrodysostosis type 2 due to a novel autosomal dominantly inherited PDE4D mutation. A 3.5-year-old boy presented with short stature, midfacial hypoplasia, severe brachydactyly, developmental delay, and behavioural problems. Laboratory investigations revealed mild thyrotropin resistance. His mother shared some characteristic features, such as midfacial hypoplasia and severe brachydactyly, but did not show short stature, intellectual disability or hormonal resistance. Genetic analysis identified the identical, novel heterozygous missense mutation of the PDE4D gene c.569C>T (p.Ser190Phe) in both patients. This case illustrates the significant phenotypic variability of acrodysostosis even within one family with identical mutations. Hence, a specific clinical diagnosis of acrodysostosis remains challenging because of great interindividual variability and a substantial overlap of the two subtypes as well as with other related Gsα-cAMP-signalling-linked disorders.
Collapse
Affiliation(s)
- Julia Hoppmann
- University of Leipzig, Hospital for Children and Adolescents, Department of Women and Child Health, Leipzig, Germany
,* Address for Correspondence: University of Leipzig, Hospital for Children and Adolescents, Department of Women and Child Health, Leipzig, Germany Phone: +49 341 972 60 00 E-mail:
| | - Julia Gesing
- University of Leipzig, Hospital for Children and Adolescents, Department of Women and Child Health, Leipzig, Germany
| | - Caroline Silve
- Université Paris-Sud Faculté de Médecine, INSERM U1169, Département de Génétique et de Biologie Moléculaire, Le Kremlin Bicêtre, France
,
Centre de Référence des Maladies Rares du Métabolisme phosphocalcique, Filiere Maladies Rares OSCAR, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Génétique et Biologie Moléculaires, Paris, France
| | - Chrystel Leroy
- Centre de Référence des Maladies Rares du Métabolisme phosphocalcique, Filiere Maladies Rares OSCAR, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Service de Génétique et Biologie Moléculaires, Paris, France
| | - Astrid Bertsche
- University of Leipzig, Hospital for Children and Adolescents, Department of Women and Child Health, Leipzig, Germany
| | - Franz Wolfgang Hirsch
- University of Leipzig, Hospital for Children and Adolescents, Department of Women and Child Health, Leipzig, Germany
| | - Wieland Kiess
- University of Leipzig, Hospital for Children and Adolescents, Department of Women and Child Health, Leipzig, Germany
| | - Roland Pfäffle
- University of Leipzig, Hospital for Children and Adolescents, Department of Women and Child Health, Leipzig, Germany
| | - Volker Schuster
- University of Leipzig, Hospital for Children and Adolescents, Department of Women and Child Health, Leipzig, Germany
| |
Collapse
|
7
|
Morris G, Walder K, McGee SL, Dean OM, Tye SJ, Maes M, Berk M. A model of the mitochondrial basis of bipolar disorder. Neurosci Biobehav Rev 2017; 74:1-20. [DOI: 10.1016/j.neubiorev.2017.01.014] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
|
8
|
Motte E, Le Stunff C, Briet C, Dumaz N, Silve C. Modulation of signaling through GPCR-cAMP-PKA pathways by PDE4 depends on stimulus intensity: Possible implications for the pathogenesis of acrodysostosis without hormone resistance. Mol Cell Endocrinol 2017; 442:1-11. [PMID: 27908835 DOI: 10.1016/j.mce.2016.11.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/25/2016] [Accepted: 11/26/2016] [Indexed: 12/30/2022]
Abstract
In acrodysostosis without hormone resistance, a disease caused by phosphodiesterase (PDE)-4D mutations, increased PDE activity leads to bone developmental defects but with normal renal responses to PTH. To identify potential mechanisms for these disparate responses, we compared the effect of PDE activity on hormone signaling through the GPCR-Gsα-cAMP-PKA pathway in cells from two lineages, HEK-293 cells stably overexpressing PTH1R (HEKpthr) and human dermal fibroblasts, including studies evaluating cAMP levels using an Epac-based BRET-sensor for cAMP (CAMYEL). For ligand-induced responses inducing strong cAMP accumulation, the inhibition of PDE4 activity resulted in relatively small further increases. In contrast, when ligand-induced cAMP accumulation was of lesser intensity, the inhibition of PDE4 had a more pronounced effect. Similar results were obtained evaluating downstream events (cellular CREB phosphorylation and CRE-luciferase activity). Thus, the ability of PDE4 to modulate signaling through GPCR-cAMP-PKA pathways can depend on the cell type and stimulus intensity.
Collapse
Affiliation(s)
- Emmanuelle Motte
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Catherine Le Stunff
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Claire Briet
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Nicolas Dumaz
- INSERM U976, Institut de Recherche sur la Peau, Hôpital Saint Louis, Paris, France
| | - Caroline Silve
- INSERM U1169, Université Paris Sud, Hôpital Bicêtre, Le Kremlin Bicêtre, France; Centre de Référence des Maladies Rares du Métabolisme du Phosphore et du Calcium, Assistance Publique Hôpitaux de Paris, Paris, France; Service de Biochimie et Génétique Moléculaires, Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Paris, France.
| |
Collapse
|
9
|
Bolger GB. The PDE4 cAMP-Specific Phosphodiesterases: Targets for Drugs with Antidepressant and Memory-Enhancing Action. ADVANCES IN NEUROBIOLOGY 2017; 17:63-102. [PMID: 28956330 DOI: 10.1007/978-3-319-58811-7_4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The PDE4 cyclic nucleotide phosphodiesterases are essential regulators of cAMP abundance in the CNS through their ability to regulate PKA activity, the phosphorylation of CREB, and other important elements of signal transduction. In pre-clinical models and in early-stage clinical trials, PDE4 inhibitors have been shown to have antidepressant and memory-enhancing activity. However, the development of clinically-useful PDE4 inhibitors for CNS disorders has been limited by variable efficacy and significant side effects. Recent structural studies have greatly enhanced our understanding of the molecular configuration of PDE4 enzymes, especially the "long" PDE4 isoforms that are abundant in the CNS. The new structural data provide a rationale for the development of a new generation of PDE4 inhibitors that specifically act on long PDE4 isoforms. These next generation PDE4 inhibitors may also be capable of targeting the interactions of select long forms with their "partner" proteins, such as RACK1, β-arrestin, and DISC1. They would therefore have the ability to affect cAMP levels in specific cellular compartments and target localized cellular functions, such as synaptic plasticity. These new agents might also be able to target PDE4 populations in select regions of the CNS that are implicated in learning and memory, affect, and cognition. Potential therapeutic uses of these agents could include affective disorders, memory enhancement, and neurogenesis.
Collapse
Affiliation(s)
- Graeme B Bolger
- Departments of Medicine and Pharmacology, University of Alabama at Birmingham, 1720 2nd Avenue South, NP 2501, Birmingham, AL, 35294-3300, USA.
| |
Collapse
|
10
|
Identification of a multifunctional docking site on the catalytic unit of phosphodiesterase-4 (PDE4) that is utilised by multiple interaction partners. Biochem J 2016; 474:597-609. [PMID: 27993970 PMCID: PMC5290487 DOI: 10.1042/bcj20160849] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/15/2016] [Accepted: 12/19/2016] [Indexed: 12/26/2022]
Abstract
Cyclic AMP (cAMP)-specific phosphodiesterase-4 (PDE4) enzymes underpin compartmentalised cAMP signalling by localising to distinct signalling complexes. PDE4 long isoforms can be phosphorylated by mitogen-activated protein kinase-activated protein kinase 2 (MK2), which attenuates activation of such enzymes through their phosphorylation by protein kinase A. Here we show that MK2 interacts directly with PDE4 long isoforms and define the sites of interaction. One is a unique site that locates within the regulatory upstream conserved region 1 (UCR1) domain and contains a core Phe141, Leu142 and Tyr143 (FLY) cluster (PDE4A5 numbering). Located with the second site is a critical core Phe693, Glu694, Phe695 (FQF) motif that is also employed in the sequestering of PDE4 long forms by an array of other signalling proteins, including the signalling scaffold β-arrestin, the tyrosyl kinase Lyn, the SUMOylation E2 ligase UBC9, the dynein regulator Lis1 (PAFAH1B1) and the protein kinase Erk. We propose that the FQF motif lies at the heart of a multifunctional docking (MFD) site located within the PDE4 catalytic unit. It is clear from our data that, as well as aiding fidelity of interaction, the MFD site confers exclusivity of binding between PDE4 and a single specific partner protein from the cohort of signalling proteins whose interaction with PDE4 involves the FQF motif.
Collapse
|
11
|
Bolger GB. RACK1 and β-arrestin2 attenuate dimerization of PDE4 cAMP phosphodiesterase PDE4D5. Cell Signal 2016; 28:706-12. [PMID: 26257302 PMCID: PMC4744576 DOI: 10.1016/j.cellsig.2015.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 08/05/2015] [Indexed: 01/14/2023]
Abstract
PDE4 family cAMP-selective cyclic nucleotide phosphodiesterases are important in the regulation of cAMP abundance in numerous systems, and thereby play an important role in the regulation of PKA and EPAC activity and the phosphorylation of CREB. We have used the yeast 2-hybrid system to demonstrate recently that long PDE4 isoforms form homodimers, consistent with data obtained recently by structural studies. The long PDE4 isoform PDE4D5 interacts selectively with β-arrestin2, implicated in the regulation of G-protein-coupled receptors and other cell signaling components, and also with the β-propeller protein RACK1. In the present study, we use 2-hybrid approaches to demonstrate that RACK1 and β-arrestin2 inhibit the dimerization of PDE4D5. We also show that serine-to-alanine mutations at PKA and ERK1/2 phosphorylation sites on PDE4D5 detectably ablate dimerization. Conversely, phospho-mimic serine-to-aspartate mutations at the MK2 and oxidative stress kinase sites ablate dimerization. Analysis of PDE4D5 that is locked into the dimeric configuration by the formation of a trans disulfide bond between Ser261 and Ser602 shows that RACK1 interacts strongly with both the monomeric and dimeric forms, but that β-arrestin2 interacts exclusively with the monomeric form. This is consistent with the concept that β-arrestin2 can preferentially recruit the monomeric, or "open," form of PDE4D5 to β2-adrenergic receptors, where it can regulate cAMP signaling.
Collapse
Affiliation(s)
- Graeme B Bolger
- Departments of Medicine and Pharmacology, University of Alabama at Birmingham, Birmingham AL 35294, USA.
| |
Collapse
|
12
|
Johanns M, Lai YC, Hsu MF, Jacobs R, Vertommen D, Van Sande J, Dumont JE, Woods A, Carling D, Hue L, Viollet B, Foretz M, Rider MH. AMPK antagonizes hepatic glucagon-stimulated cyclic AMP signalling via phosphorylation-induced activation of cyclic nucleotide phosphodiesterase 4B. Nat Commun 2016; 7:10856. [PMID: 26952277 PMCID: PMC4786776 DOI: 10.1038/ncomms10856] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 01/27/2016] [Indexed: 11/14/2022] Open
Abstract
Biguanides such as metformin have previously been shown to antagonize hepatic glucagon-stimulated cyclic AMP (cAMP) signalling independently of AMP-activated protein kinase (AMPK) via direct inhibition of adenylate cyclase by AMP. Here we show that incubation of hepatocytes with the small-molecule AMPK activator 991 decreases glucagon-stimulated cAMP accumulation, cAMP-dependent protein kinase (PKA) activity and downstream PKA target phosphorylation. Moreover, incubation of hepatocytes with 991 increases the Vmax of cyclic nucleotide phosphodiesterase 4B (PDE4B) without affecting intracellular adenine nucleotide concentrations. The effects of 991 to decrease glucagon-stimulated cAMP concentrations and activate PDE4B are lost in hepatocytes deleted for both catalytic subunits of AMPK. PDE4B is phosphorylated by AMPK at three sites, and by site-directed mutagenesis, Ser304 phosphorylation is important for activation. In conclusion, we provide a new mechanism by which AMPK antagonizes hepatic glucagon signalling via phosphorylation-induced PDE4B activation. The diabetes drug Metformin decreases hepatic glucose production and activates AMP-activated protein kinase (AMPK). Here the authors provide evidence that AMPK activation antagonizes glucagon signalling by activating PDE4B, lowering cAMP levels and decreasing PKA activation.
Collapse
Affiliation(s)
- M Johanns
- Université catholique de Louvain and de Duve Institute, Avenue Hippocrate, 75, 1200 Brussels, Belgium
| | - Y-C Lai
- Université catholique de Louvain and de Duve Institute, Avenue Hippocrate, 75, 1200 Brussels, Belgium
| | - M-F Hsu
- Université catholique de Louvain and de Duve Institute, Avenue Hippocrate, 75, 1200 Brussels, Belgium
| | - R Jacobs
- Université catholique de Louvain and de Duve Institute, Avenue Hippocrate, 75, 1200 Brussels, Belgium
| | - D Vertommen
- Université catholique de Louvain and de Duve Institute, Avenue Hippocrate, 75, 1200 Brussels, Belgium
| | - J Van Sande
- Faculté de Médecine, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070 Brussels, Belgium
| | - J E Dumont
- Faculté de Médecine, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070 Brussels, Belgium
| | - A Woods
- Cellular Stress Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, DuCane Road, London W12 0NN, UK
| | - D Carling
- Cellular Stress Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, DuCane Road, London W12 0NN, UK
| | - L Hue
- Université catholique de Louvain and de Duve Institute, Avenue Hippocrate, 75, 1200 Brussels, Belgium
| | - B Viollet
- INSERM U1016, Institut Cochin, 75014 Paris, France.,CNRS UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - M Foretz
- INSERM U1016, Institut Cochin, 75014 Paris, France.,CNRS UMR8104, 75014 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - M H Rider
- Université catholique de Louvain and de Duve Institute, Avenue Hippocrate, 75, 1200 Brussels, Belgium
| |
Collapse
|
13
|
Böttcher R, Henderson DJP, Dulla K, van Strijp D, Waanders LF, Tevz G, Lehman ML, Merkle D, van Leenders GJLH, Baillie GS, Jenster G, Houslay MD, Hoffmann R. Human phosphodiesterase 4D7 (PDE4D7) expression is increased in TMPRSS2-ERG-positive primary prostate cancer and independently adds to a reduced risk of post-surgical disease progression. Br J Cancer 2016; 113:1502-11. [PMID: 26575822 PMCID: PMC4815894 DOI: 10.1038/bjc.2015.335] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/14/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023] Open
Abstract
Background: There is an acute need to uncover biomarkers that reflect the molecular pathologies, underpinning prostate cancer progression and poor patient outcome. We have previously demonstrated that in prostate cancer cell lines PDE4D7 is downregulated in advanced cases of the disease. To investigate further the prognostic power of PDE4D7 expression during prostate cancer progression and assess how downregulation of this PDE isoform may affect disease outcome, we have examined PDE4D7 expression in physiologically relevant primary human samples. Methods: About 1405 patient samples across 8 publically available qPCR, Affymetrix Exon 1.0 ST arrays and RNA sequencing data sets were screened for PDE4D7 expression. The TMPRSS2-ERG gene rearrangement status of patient samples was determined by transformation of the exon array and RNA seq expression data to robust z-scores followed by the application of a threshold >3 to define a positive TMPRSS2-ERG gene fusion event in a tumour sample. Results: We demonstrate that PDE4D7 expression positively correlates with primary tumour development. We also show a positive association with the highly prostate cancer-specific gene rearrangement between TMPRSS2 and the ETS transcription factor family member ERG. In addition, we find that in primary TMPRSS2-ERG-positive tumours PDE4D7 expression is significantly positively correlated with low-grade disease and a reduced likelihood of progression after primary treatment. Conversely, PDE4D7 transcript levels become significantly decreased in castration resistant prostate cancer (CRPC). Conclusions: We further characterise and add physiological relevance to PDE4D7 as a novel marker that is associated with the development and progression of prostate tumours. We propose that the assessment of PDE4D7 levels may provide a novel, independent predictor of post-surgical disease progression.
Collapse
Affiliation(s)
- R Böttcher
- Department of Urology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - D J P Henderson
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow G12 8TA, Scotland
| | - K Dulla
- Departments of Oncology Solutions and Precision Diagnostics, Philips Research Europe, Eindhoven 5656 AE, The Netherlands
| | - D van Strijp
- Departments of Oncology Solutions and Precision Diagnostics, Philips Research Europe, Eindhoven 5656 AE, The Netherlands
| | - L F Waanders
- Departments of Oncology Solutions and Precision Diagnostics, Philips Research Europe, Eindhoven 5656 AE, The Netherlands
| | - G Tevz
- Departments of Oncology Solutions and Precision Diagnostics, Philips Research Europe, Eindhoven 5656 AE, The Netherlands.,Australian Prostate Cancer Research Centre-Institute of Health and Biomedical Innovation, University of Technology, and Translational Research Institute, Brisbane, Queensland 4102, Australia
| | - M L Lehman
- Australian Prostate Cancer Research Centre-Institute of Health and Biomedical Innovation, University of Technology, and Translational Research Institute, Brisbane, Queensland 4102, Australia
| | - D Merkle
- Departments of Oncology Solutions and Precision Diagnostics, Philips Research Europe, Eindhoven 5656 AE, The Netherlands
| | - G J L H van Leenders
- Department of Pathology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - G S Baillie
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow G12 8TA, Scotland
| | - G Jenster
- Department of Urology, Erasmus Medical Center, Rotterdam 3000 CA, The Netherlands
| | - M D Houslay
- Institute of Pharmaceutical Science, King's College London, London WC2R 2LS, UK
| | - R Hoffmann
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow G12 8TA, Scotland.,Departments of Oncology Solutions and Precision Diagnostics, Philips Research Europe, Eindhoven 5656 AE, The Netherlands
| |
Collapse
|
14
|
PDE4D phosphorylation: A coincidence detector integrating multiple signaling pathways. Cell Signal 2015; 28:719-24. [PMID: 26562185 DOI: 10.1016/j.cellsig.2015.11.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/02/2015] [Indexed: 11/24/2022]
Abstract
In Eukaryotes, more than 100 different phosphodiesterase (PDE) proteins serve to fine-tune cyclic nucleotide (cAMP and cGMP) signals and contribute to specificity of signaling. In mammals, PDEs are divided into 11 families, of which PDE4 represents the largest family. Four genes (pde4a, pde4b, pde4c and pde4d) encode for this class of enzymes in mammals and give rise to more than 20 variants. Within this family of genes, PDE4D was discovered on the basis of its regulatory properties and its induction by hormones and cAMP. PDE4D has often been used as the prototype PDE4 and large body of work has been generated on the biochemical, pharmacological, and physiological properties of this enzyme. This review covers the regulation of PDE4D by phosphorylation, the impact of this regulation in the context of the structure of this protein, and the functional consequences of this complex pattern of posttranslational modifications.
Collapse
|
15
|
Uncovering the function of Disrupted in Schizophrenia 1 through interactions with the cAMP phosphodiesterase PDE4: Contributions of the Houslay lab to molecular psychiatry. Cell Signal 2015; 28:749-52. [PMID: 26432168 DOI: 10.1016/j.cellsig.2015.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022]
Abstract
Nearly 10years ago the laboratory of Miles Houslay was part of a collaboration which identified and characterized the interaction between Disrupted in Schizophrenia 1 and phosphodiesterase type 4. This work has had significant impact on our thinking of psychiatric illness causation and the potential for therapeutics.
Collapse
|
16
|
Lundegaard PR, Anastasaki C, Grant NJ, Sillito RR, Zich J, Zeng Z, Paranthaman K, Larsen AP, Armstrong JD, Porteous DJ, Patton EE. MEK Inhibitors Reverse cAMP-Mediated Anxiety in Zebrafish. ACTA ACUST UNITED AC 2015; 22:1335-46. [PMID: 26388333 PMCID: PMC4623357 DOI: 10.1016/j.chembiol.2015.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 08/11/2015] [Accepted: 08/14/2015] [Indexed: 12/14/2022]
Abstract
Altered phosphodiesterase (PDE)-cyclic AMP (cAMP) activity is frequently associated with anxiety disorders, but current therapies act by reducing neuronal excitability rather than targeting PDE-cAMP-mediated signaling pathways. Here, we report the novel repositioning of anti-cancer MEK inhibitors as anxiolytics in a zebrafish model of anxiety-like behaviors. PDE inhibitors or activators of adenylate cyclase cause behaviors consistent with anxiety in larvae and adult zebrafish. Small-molecule screening identifies MEK inhibitors as potent suppressors of cAMP anxiety behaviors in both larvae and adult zebrafish, while causing no anxiolytic behavioral effects on their own. The mechanism underlying cAMP-induced anxiety is via crosstalk to activation of the RAS-MAPK signaling pathway. We propose that targeting crosstalk signaling pathways can be an effective strategy for mental health disorders, and advance the repositioning of MEK inhibitors as behavior stabilizers in the context of increased cAMP.
Collapse
Affiliation(s)
- Pia R Lundegaard
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; Department of Biomedical Sciences, Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Corina Anastasaki
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Nicola J Grant
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Rowland R Sillito
- Actual Analytics Ltd, 2.05 Wilkie Building, 22-23 Teviot Row, Edinburgh EH8 9AG, UK
| | - Judith Zich
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Zhiqiang Zeng
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Karthika Paranthaman
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Anders Peter Larsen
- Department of Biomedical Sciences, Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, 2200 Copenhagen, Denmark
| | - J Douglas Armstrong
- Actual Analytics Ltd, 2.05 Wilkie Building, 22-23 Teviot Row, Edinburgh EH8 9AG, UK; School of Informatics, Institute for Adaptive and Neural Computation, Informatics Forum, University of Edinburgh, Edinburgh EH8 9AB, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.
| | - E Elizabeth Patton
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK; Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh EH4 2XU, UK; Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.
| |
Collapse
|
17
|
Vandame P, Spriet C, Trinel D, Gelaude A, Caillau K, Bompard C, Biondi E, Bodart JF. The spatio-temporal dynamics of PKA activity profile during mitosis and its correlation to chromosome segregation. Cell Cycle 2015; 13:3232-40. [PMID: 25485503 DOI: 10.4161/15384101.2014.950907] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The cyclic adenosine monophosphate dependent kinase protein (PKA) controls a variety of cellular processes including cell cycle regulation. Here, we took advantages of genetically encoded FRET-based biosensors, using an AKAR-derived biosensor to characterize PKA activity during mitosis in living HeLa cells using a single-cell approach. We measured PKA activity changes during mitosis. HeLa cells exhibit a substantial increase during mitosis, which ends with telophase. An AKAREV T>A inactive form of the biosensor and H89 inhibitor were used to ascertain for the specificity of the PKA activity measured. On a spatial point of view, high levels of activity near to chromosomal plate during metaphase and anaphase were detected. By using the PKA inhibitor H89, we assessed the role of PKA in the maintenance of a proper division phenotype. While this treatment in our hands did not impaired cell cycle progression in a drastic manner, inhibition of PKA leads to a dramatic increase in chromososme misalignement on the spindle during metaphase that could result in aneuploidies. Our study emphasizes the insights that can be gained with genetically encoded FRET-based biosensors, which enable to overcome the shortcomings of classical methologies and unveil in vivo PKA spatiotemporal profiles in HeLa cells.
Collapse
Affiliation(s)
- Pauline Vandame
- a Laboratoire de Régulation des Signaux de division; EA4479; Université Lille1; Université Lille Nord de France; Villeneuve d'Ascq, France Institut Fédératif de Recherche (IFR)147; Site de Recherche Intégré en Cancérologie (SIRIC) ONCOLILLE
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Cell division relies on coordinated regulation of the cell cycle. A process including a well-defined series of strictly regulated molecular mechanisms involving cyclin-dependent kinases, retinoblastoma protein, and polo-like kinases. Dysfunctions in cell cycle regulation are associated with disease such as cancer, diabetes, and neurodegeneration. Compartmentalization of cellular signaling is a common strategy used to ensure the accuracy and efficiency of cellular responses. Compartmentalization of intracellular signaling is maintained by scaffolding proteins, such as A-kinase anchoring proteins (AKAPs). AKAPs are characterized by their ability to anchor the regulatory subunits of protein kinase A (PKA), and thereby achieve guidance to different cellular locations via various targeting domains. Next to PKA, AKAPs also associate with several other signaling elements including receptors, ion channels, protein kinases, phosphatases, small GTPases, and phosphodiesterases. Taking the amount of possible AKAP signaling complexes and their diverse localization into account, it is rational to believe that such AKAP-based complexes regulate several critical cellular events of the cell cycle. In fact, several AKAPs are assigned as tumor suppressors due to their vital roles in cell cycle regulation. Here, we first briefly discuss the most important players of cell cycle progression. After that, we will review our recent knowledge of AKAPs linked to the regulation and progression of the cell cycle, with special focus on AKAP12, AKAP8, and Ezrin. At last, we will discuss this specific AKAP subset in relation to diseases with focus on a diverse subset of cancer.
Collapse
Affiliation(s)
- B Han
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands. .,Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands.
| | - W J Poppinga
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| | - M Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| |
Collapse
|
19
|
Wang L, Burmeister BT, Johnson KR, Baillie GS, Karginov AV, Skidgel RA, O'Bryan JP, Carnegie GK. UCR1C is a novel activator of phosphodiesterase 4 (PDE4) long isoforms and attenuates cardiomyocyte hypertrophy. Cell Signal 2015; 27:908-22. [PMID: 25683917 DOI: 10.1016/j.cellsig.2015.02.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 02/03/2015] [Accepted: 02/05/2015] [Indexed: 01/21/2023]
Abstract
Hypertrophy increases the risk of heart failure and arrhythmia. Prevention or reversal of the maladaptive hypertrophic phenotype has thus been proposed to treat heart failure. Chronic β-adrenergic receptor (β-AR) stimulation induces cardiomyocyte hypertrophy by elevating 3',5'-cyclic adenosine monophosphate (cAMP) levels and activating downstream effectors such protein kinase A (PKA). Conversely, hydrolysis of cAMP by phosphodiesterases (PDEs) spatiotemporally restricts cAMP signaling. Here, we demonstrate that PDE4, but not PDE3, is critical in regulating cardiomyocyte hypertrophy, and may represent a potential target for preventing maladaptive hypertrophy. We identify a sequence within the upstream conserved region 1 of PDE4D, termed UCR1C, as a novel activator of PDE4 long isoforms. UCR1C activates PDE4 in complex with A-kinase anchoring protein (AKAP)-Lbc resulting in decreased PKA signaling facilitated by AKAP-Lbc. Expression of UCR1C in cardiomyocytes inhibits hypertrophy in response to chronic β-AR stimulation. This effect is partially due to inhibition of nuclear PKA activity, which decreases phosphorylation of the transcription factor cAMP response element-binding protein (CREB). In conclusion, PDE4 activation by UCR1C attenuates cardiomyocyte hypertrophy by specifically inhibiting nuclear PKA activity.
Collapse
Affiliation(s)
- Li Wang
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA
| | - Brian T Burmeister
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA
| | - Keven R Johnson
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA
| | - George S Baillie
- Institute of Cardiovascular and Medical Science, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G128QQ, Scotland, United Kingdom
| | - Andrei V Karginov
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA; University of Illinois Cancer Center, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA
| | - Randal A Skidgel
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA
| | - John P O'Bryan
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA; University of Illinois Cancer Center, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA; Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA; Jessie Brown VA Medical Center, 820 S Damen Ave, Chicago, IL 60612, USA.
| | - Graeme K Carnegie
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, E403 MSB, 835 South Wolcott Avenue, Chicago, IL 60612, USA
| |
Collapse
|
20
|
Bolger GB, Dunlop AJ, Meng D, Day JP, Klussmann E, Baillie GS, Adams DR, Houslay MD. Dimerization of cAMP phosphodiesterase-4 (PDE4) in living cells requires interfaces located in both the UCR1 and catalytic unit domains. Cell Signal 2014; 27:756-69. [PMID: 25546709 PMCID: PMC4371794 DOI: 10.1016/j.cellsig.2014.12.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 12/16/2014] [Indexed: 02/08/2023]
Abstract
PDE4 family cAMP phosphodiesterases play a pivotal role in determining compartmentalised cAMP signalling through targeted cAMP breakdown. Expressing the widely found PDE4D5 isoform, as both bait and prey in a yeast 2-hybrid system, we demonstrated interaction consistent with the notion that long PDE4 isoforms form dimers. Four potential dimerization sites were uncovered using a scanning peptide array approach, where a recombinant purified PDE4D5 fusion protein was used to probe a 25-mer library of overlapping peptides covering the entire PDE4D5 sequence. Key residues involved in PDE4D5 dimerization were defined using a site-directed mutagenesis programme directed by an alanine scanning peptide array approach. Critical residues stabilising PDE4D5 dimerization were defined within the regulatory UCR1 region found in long, but not short, PDE4 isoforms, namely the Arg173, Asn174 and Asn175 (DD1) cluster. Disruption of the DD1 cluster was not sufficient, in itself, to destabilise PDE4D5 homodimers. Instead, disruption of an additional interface, located on the PDE4 catalytic unit, was also required to convert PDE4D5 into a monomeric form. This second dimerization site on the conserved PDE4 catalytic unit is dependent upon a critical ion pair interaction. This involves Asp463 and Arg499 in PDE4D5, which interact in a trans fashion involving the two PDE4D5 molecules participating in the homodimer. PDE4 long isoforms adopt a dimeric state in living cells that is underpinned by two key contributory interactions, one involving the UCR modules and one involving an interface on the core catalytic domain. We propose that short forms do not adopt a dimeric configuration because, in the absence of the UCR1 module, residual engagement of the remaining core catalytic domain interface provides insufficient free energy to drive dimerization. The functioning of PDE4 long and short forms is thus poised to be inherently distinct due to this difference in quaternary structure. In a yeast 2-hybrid system we show that long PDE4 isoforms dimerize. Scanning peptide array and mutagenesis located two dimerization surfaces. One surface maps to the regulatory UCR1 region found only in long forms. A second locates to the core catalytic domain. PDE4 long and short forms differ in quaternary structure.
Collapse
Affiliation(s)
- Graeme B Bolger
- Departments of Medicine and Pharmacology, University of Alabama, Birmingham, AL 35294, USA
| | - Allan J Dunlop
- Institute of Cardiovascular and Medical Science, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Dong Meng
- Institute of Cardiovascular and Medical Science, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Jon P Day
- Institute of Cardiovascular and Medical Science, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Enno Klussmann
- Max Delbrueck Center for Molecular Medicine, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - George S Baillie
- Institute of Cardiovascular and Medical Science, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - David R Adams
- Institute of Chemical Sciences, Heriot-Watt University, Riccarton, Edinburgh EH14 4AS, Scotland, United Kingdom
| | - Miles D Houslay
- Institute of Pharmaceutical Sciences, King's College London, London SE1 9NH, United Kingdom.
| |
Collapse
|
21
|
He N, Kim N, Song M, Park C, Kim S, Park EY, Yim HY, Kim K, Park JH, Kim KI, Zhang F, Mills GB, Yoon S. Integrated analysis of transcriptomes of cancer cell lines and patient samples reveals STK11/LKB1-driven regulation of cAMP phosphodiesterase-4D. Mol Cancer Ther 2014; 13:2463-73. [PMID: 25122068 DOI: 10.1158/1535-7163.mct-14-0297] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The recent proliferation of data on large collections of well-characterized cancer cell lines linked to therapeutic drug responses has made it possible to identify lineage- and mutation-specific transcriptional markers that can help optimize implementation of anticancer agents. Here, we leverage these resources to systematically investigate the presence of mutation-specific transcription markers in a wide variety of cancer lineages and genotypes. Sensitivity and specificity of potential transcriptional biomarkers were simultaneously analyzed in 19 cell lineages grouped into 228 categories based on the mutational genotypes of 12 cancer-related genes. Among a total of 1,455 category-specific expression patterns, the expression of cAMP phosphodiesterase-4D (PDE4D) with 11 isoforms, one of the PDE4(A-D) subfamilies, was predicted to be regulated by a mutant form of serine/threonine kinase 11 (STK11)/liver kinase B1 (LKB1) present in lung cancer. STK11/LKB1 is the primary upstream kinase of adenine monophosphate-activated protein kinase (AMPK). Subsequently, we found that the knockdown of PDE4D gene expression inhibited proliferation of STK11-mutated lung cancer lines. Furthermore, challenge with a panel of PDE4-specific inhibitors was shown to selectively reduce the growth of STK11-mutated lung cancer lines. Thus, we show that multidimensional analysis of a well-characterized large-scale panel of cancer cell lines provides unprecedented opportunities for the identification of unexpected oncogenic mechanisms and mutation-specific drug targets.
Collapse
Affiliation(s)
- Ningning He
- Center for Advanced Bioinformatics and Systems Medicine, Sookmyung Women's University, Seoul, Republic of Korea. Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Nayoung Kim
- Center for Advanced Bioinformatics and Systems Medicine, Sookmyung Women's University, Seoul, Republic of Korea. Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Mee Song
- Center for Advanced Bioinformatics and Systems Medicine, Sookmyung Women's University, Seoul, Republic of Korea
| | - Choa Park
- Center for Advanced Bioinformatics and Systems Medicine, Sookmyung Women's University, Seoul, Republic of Korea
| | - Somin Kim
- Center for Advanced Bioinformatics and Systems Medicine, Sookmyung Women's University, Seoul, Republic of Korea
| | - Eun Young Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Hwa Young Yim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Kyunga Kim
- Department of Statistics, Sookmyung Women's University, Seoul, Republic of Korea
| | - Jong Hoon Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Keun Il Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Fan Zhang
- Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gordon B Mills
- Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sukjoon Yoon
- Center for Advanced Bioinformatics and Systems Medicine, Sookmyung Women's University, Seoul, Republic of Korea. Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Heterozygous mutations in cyclic AMP phosphodiesterase-4D (PDE4D) and protein kinase A (PKA) provide new insights into the molecular pathology of acrodysostosis. Cell Signal 2014; 26:2446-59. [PMID: 25064455 DOI: 10.1016/j.cellsig.2014.07.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 07/16/2014] [Accepted: 07/16/2014] [Indexed: 12/21/2022]
Abstract
Acrodysostosis without hormone resistance is a rare skeletal disorder characterized by brachydactyly, nasal hypoplasia, mental retardation and occasionally developmental delay. Recently, loss-of-function mutations in the gene encoding cAMP-hydrolyzing phosphodiesterase-4D (PDE4D) have been reported to cause this rare condition but the pathomechanism has not been fully elucidated. To understand the pathogenetic mechanism of PDE4D mutations, we conducted 3D modeling studies to predict changes in the binding efficacy of cAMP to the catalytic pocket in PDE4D mutants. Our results indicated diminished enzyme activity in the two mutants we analyzed (Gly673Asp and Ile678Thr; based on PDE4D4 residue numbering). Ectopic expression of PDE4D mutants in HEK293 cells demonstrated this reduction in activity, which was identified by increased cAMP levels. However, the cells from an acrodysostosis patient showed low cAMP accumulation, which resulted in a decrease in the phosphorylated cAMP Response Element-Binding Protein (pCREB)/CREB ratio. The reason for this discrepancy was due to a compensatory increase in expression levels of PDE4A and PDE4B isoforms, which accounted for the paradoxical decrease in cAMP levels in the patient cells expressing mutant isoforms with a lowered PDE4D activity. Skeletal radiographs of 10-week-old knockout (KO) rats showed that the distal part of the forelimb was shorter than in wild-type (WT) rats and that all the metacarpals and phalanges were also shorter in KO, as the name acrodysostosis implies. Like the G-protein α-stimulatory subunit and PRKAR1A, PDE4D critically regulates the cAMP signal transduction pathway and influences bone formation in a way that activity-compromising PDE4D mutations can result in skeletal dysplasia. We propose that specific inhibitory PDE4D mutations can lead to the molecular pathology of acrodysostosis without hormone resistance but that the pathological phenotype may well be dependent on an over-compensatory induction of other PDE4 isoforms that can be expected to be targeted to different signaling complexes and exert distinct effects on compartmentalized cAMP signaling.
Collapse
|