1
|
Zhao Q, Liow JS, Jee JE, Montero Santamaria J, Pamie-George M, Morse C, Wu S, Zoghbi SS, Kim SW, Innis RB, Pike VW, Telu S. [ 11C]ZTP-1: An Effective Short-Lived Radioligand for PET of Rat and Monkey Brain Phosphodiesterase Type 4 Subtype B. J Nucl Med 2025:jnumed.124.269159. [PMID: 40341096 DOI: 10.2967/jnumed.124.269159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/31/2025] [Indexed: 05/10/2025] Open
Abstract
Phosphodiesterase type 4 subtype B (PDE4B) selectively hydrolyzes cyclic adenosine monophosphate to enact numerous downstream signaling events. PDE4B is widely expressed in the brain and is implicated in several neuropsychiatric disorders. Moreover, PDE4B inhibition shows antiinflammatory and antidepressant-like effects in animal studies. [18F]PF-06445974 has been developed to image human brain PDE4B using PET, thereby providing a tool for pathophysiologic studies and drug development. However, a radioligand labeled with shorter-lived 11C would be an alternative for studies that require more than 1 administration into the same imaging subject on a single day. Methods: 8-Cyclopropyl-10-(3,5-difluoro-4-(methoxy)phenyl)-7,8-dihydropyrido[2',3':4,5]pyrrolo[1,2-a]pyrazin-9(6H)-1 (ZTP-1) was identified as possessing many favorable properties for development as a 11C-labeled PET radioligand, including high PDE4B inhibitory potency, moderate computed lipophilicity, and a methoxy group as a potential labeling site. Here, [11C]ZTP-1 was readily obtained by 11C methylation of a synthesized O-desmethyl precursor. PET imaging of rat and rhesus monkey brains was performed with [11C]ZTP-1 at baseline and after administration of PDE4B- and PDE4D-selective inhibitors. Radiometabolite profiles for [11C]ZTP-1 were also determined ex vivo in rat plasma and brains. Results: [11C]ZTP-1 was obtained in a high activity yield and with high molar activity. Rat and monkey PET imaging showed high whole-brain radioactivity uptake with subsequent gradual washout. Challenge experiments verified a high and PDE4B-selective PET signal in rat and monkey brains. Ex vivo rat brain uptake of [11C]ZTP-1 showed less than 1% radiometabolite contamination at 30 min. Total distribution volume measures in monkey brains quickly reached stability. Conclusion: [11C]ZTP-1 is a promising, shorter-lived alternative to [18F]PF-06445974 for quantifying brain PDE4B in rodents and nonhuman primates with PET and warrants further investigation in humans.
Collapse
Affiliation(s)
- Qunchao Zhao
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Joo Eun Jee
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Jose Montero Santamaria
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Matilah Pamie-George
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Cheryl Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Shawn Wu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Sung Won Kim
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
2
|
Banerjee A, Thekkekkara D, Manjula SN, Nair SP, Lalitha MS. Correlation of autophagy and Alzheimer's disease with special emphasis on the role of phosphodiesterase-4. 3 Biotech 2025; 15:139. [PMID: 40292249 PMCID: PMC12018668 DOI: 10.1007/s13205-025-04306-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Autophagy disruption is important in Alzheimer's disease (AD) as it prevents misfolded proteins from being removed, which leads to the accumulation of amyloid plaques and neurofibrillary tangles (NFTs). Restoring autophagy improves neuronal survival and cognitive function, according to experimental models. In AD models, mTOR inhibition and AMPK activation enhance synaptic plasticity and lessen learning deficits. Inhibitors of phosphodiesterase-4 (PDE4) improve cognition and reduce neuroinflammation via altering cyclic adenosine monophosphate (cAMP) transmission. Furthermore, autophagic-lysosomal clearance is encouraged by upregulating transcription factor EB (TFEB), which lessens the pathogenic damage linked to AD. These results point to autophagy modification as a promising therapeutic approach, with the mTOR, AMPK, cAMP, and TFEB pathways being possible targets for drugs. Though much evidence is based on animal studies, these findings provide valuable insights into autophagy's role in AD pathology, offering promising directions for future research and drug development.
Collapse
Affiliation(s)
- Aniruddha Banerjee
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Salini P. Nair
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Mankala Sree Lalitha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| |
Collapse
|
3
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Bhattacharya A, Turkalj L, Manzini MC. The promise of cyclic AMP modulation to restore cognitive function in neurodevelopmental disorders. Curr Opin Neurobiol 2025; 90:102966. [PMID: 39740265 DOI: 10.1016/j.conb.2024.102966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Cyclic AMP (cAMP) is a key regulator of synaptic function and is dysregulated in both neurodevelopmental (NDD) and neurodegenerative disorders. Due to the ease of diffusion and promiscuity of downstream effectors, cAMP signaling is restricted within spatiotemporal domains to localize activation. Among the best-studied mechanisms is the feedback inhibition of cAMP-dependent protein kinase (PKA) activity by phosphodiesterases 4 (PDE4s) at synapses controlling neuronal plasticity, which is largely regulated by PDE4D. In fact, genetic variants in genes for multiple PKA subunits and PDE4D lead to NDDs. Here, we discuss the rationale for choosing PDE4D as a candidate for the design of selective allosteric inhibitors and the recent advances in clinical trials. These new compounds improve cognitive function in preclinical animal models due to improved selectivity and more physiological inhibition of the active enzyme. We also discuss opportunities for better understanding of PDE4D function in general, and for the development of next-generation inhibitors.
Collapse
Affiliation(s)
- Aniket Bhattacharya
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, 08901, USA
| | - Luka Turkalj
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, 08901, USA
| | - M Chiara Manzini
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, 08901, USA.
| |
Collapse
|
5
|
Schepers M, Vangansewinkel T, Libberecht K, Jeurissen H, Jacobs D, Piccart E, Prior R, Ricciarelli R, Brullo C, Fedele E, Bruno O, Prickaerts J, Lambrichts I, Van Den Bosch L, Vanmierlo T, Wolfs E. Phosphodiesterase 4D inhibition improves the functional and molecular outcome in a mouse and human model of Charcot Marie Tooth disease 1 A. Biomed Pharmacother 2025; 183:117828. [PMID: 39823724 DOI: 10.1016/j.biopha.2025.117828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/20/2025] Open
Abstract
Charcot-Marie-Tooth disease type 1A (CMT1A) is an inherited peripheral neuropathy caused by a duplication of the peripheral myelin protein 22 (PMP22) gene. It is primarily marked by Schwann cell dedifferentiation and demyelination, leading to motor and sensory deficits. Cyclic adenosine monophosphate (cAMP) is crucial for Schwann cell differentiation and maturation. Therefore, increasing cAMP by inhibiting its degraders, phosphodiesterases (PDE), is a potential therapeutic strategy for CMT1A. This study investigated the therapeutic potential of the specific PDE4D inhibitor Gebr32a using the C3-PMP22 mouse model for CMT1A and patient-induced Pluripotent Stem Cell (iPSC)-derived Schwann cells. C3-PMP22 mice, injected subcutaneously with Gebr32a twice a day for 10 weeks, showed significantly increased nerve conduction in sciatic nerves compared to vehicle-injected controls, indicating improved myelination. Additionally, Gebr32a-treated C3-PMP22 mice exhibited improved sensorimotor functions. Grip strength analysis revealed significantly increased strength in all limbs of Gebr32a-treated C3-PMP22 mice. Post-mortem histological and ultrastructural analysis confirmed enhanced myelination in the sciatic nerve of treated mice compared to controls. In primary mouse CMT1A Schwann cells, Gebr32a dose-dependently increased the expression of pro-myelinating genes such as oct6, Krox20, Mbp, Mpz, and Plp, while downregulating the dedifferentiation marker c-Jun and human PMP22. Similar effects on gene expression were observed in iPSC-derived Schwann cells from a CMT1A patient, highlighting the clinical relevance of our findings. In conclusion, inhibition of PDE4D with Gebr32a improves the functional and molecular outcomes in mouse and human models of CMT1A, highlighting its potential as a new therapeutic strategy for CMT1A disease management.
Collapse
Affiliation(s)
- Melissa Schepers
- NIC&R - Neuro-Immune Connection & Repair, BIOMED, Department of Neuroscience, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience - Division Translational Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Tim Vangansewinkel
- Laboratory for Functional Imaging & Research on Stem Cells, BIOMED, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven , Belgium
| | - Karen Libberecht
- Laboratory for Functional Imaging & Research on Stem Cells, BIOMED, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven , Belgium
| | - Hanne Jeurissen
- Laboratory for Functional Imaging & Research on Stem Cells, BIOMED, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Darren Jacobs
- NIC&R - Neuro-Immune Connection & Repair, BIOMED, Department of Neuroscience, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; Laboratory for Functional Imaging & Research on Stem Cells, BIOMED, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Elisabeth Piccart
- NIC&R - Neuro-Immune Connection & Repair, BIOMED, Department of Neuroscience, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Robert Prior
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven , Belgium; Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Roberta Ricciarelli
- IRCCS Ospedale Policlinico San Martino, Genova 16100, Italy; Department of Experimental Medicine, Section of General Pathology, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Ernesto Fedele
- IRCCS Ospedale Policlinico San Martino, Genova 16100, Italy; Department of Pharmacy, Section of Pharmacology and Toxicology, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Olga Bruno
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | | | - Ivo Lambrichts
- Laboratory for Functional Imaging & Research on Stem Cells, BIOMED, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven - University of Leuven, Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven , Belgium
| | - Tim Vanmierlo
- NIC&R - Neuro-Immune Connection & Repair, BIOMED, Department of Neuroscience, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience - Division Translational Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Esther Wolfs
- Laboratory for Functional Imaging & Research on Stem Cells, BIOMED, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
6
|
Kumari S, Bagri K, Deshmukh R. Connecting dots: Preclinical foundations to clinical realities of PDE4 inhibitors in Alzheimer's disease. Inflammopharmacology 2025; 33:593-603. [PMID: 39808238 DOI: 10.1007/s10787-024-01638-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Alzheimer's Disease (AD), a progressive and age-associated neurodegenerative disorder, is primarily characterized by amyloid-beta (Aβ) plaques and neurofibrillary tangles. Despite advances in targeting Aβ-mediated neuronal damage with anti-Aβ antibodies, these treatments provide only symptomatic relief and fail to address the multifactorial pathology of the disease. This necessitates the exploration of novel therapeutic approaches and a deeper understanding of molecular signaling mechanisms underlying AD. Phosphodiesterases (PDEs), particularly Phosphodiesterase 4 (PDE4), play a pivotal role in regulating cyclic adenosine monophosphate (cAMP), a key molecule involved in memory consolidation and cognitive function. PDE4 inhibitors have demonstrated potential in enhancing memory and cognition in preclinical models of AD by modulating cAMP signaling. However, their clinical translation has been limited due to challenges such as adverse effects, narrow therapeutic windows, and low specificity in mechanism of action. This review bridges the gap between preclinical discoveries and clinical applications of PDE4 inhibitors in AD. It highlights preclinical evidence supporting the neuroprotective and anti-inflammatory effects of PDE4 inhibitors while addressing challenges in their clinical development, including issues of safety, efficacy, and disease-specific targeting. By integrating findings from both preclinical and clinical studies, we provide a comprehensive understanding of the therapeutic potential of PDE4 inhibitors in AD. Furthermore, this review outlines future research directions aimed at optimizing PDE4 inhibition strategies for AD treatment, offering a roadmap to translate foundational insights into clinical realities.
Collapse
Affiliation(s)
- Shilpa Kumari
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, Punjab, India
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Kajal Bagri
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, Punjab, India
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Rahul Deshmukh
- Department of Pharmacology, Central University of Punjab, Bathinda, 151001, Punjab, India.
| |
Collapse
|
7
|
Desouky MA, Michel HE, Elsherbiny DA, George MY. Recent pharmacological insights on abating toxic protein species burden in neurological disorders: Emphasis on 26S proteasome activation. Life Sci 2024; 359:123206. [PMID: 39489397 DOI: 10.1016/j.lfs.2024.123206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/30/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Protein homeostasis (proteostasis) refers to the plethora of mechanisms that safeguard the proper folding of the newly synthesized proteins. It entails various intricately regulated cues that demolish the toxic protein species to prevent their aggregation. The ubiquitin-proteasome system (UPS) is recognized as a salient protein degradation system, with a substantial role in maintaining proteostasis. However, under certain circumstances the protein degradation capacity of the UPS is overwhelmed, leading to the accumulation of misfolded proteins. Several neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, Huntington disease, and amyotrophic lateral sclerosis are characterized with the presence of protein aggregates and proteinopathy. Accordingly, enhancing the 26S proteasome degradation activity might delineate a pioneering approach in targeting various proteotoxic disorders. Regrettably, the exact molecular approaches that enhance the proteasomal activity are still not fully understood. Therefore, this review aimed to underscore several signaling cascades that might restore the degradation capacity of this molecular machine. In this review, we discuss the different molecular components of the UPS and how 26S proteasomes are deleteriously affected in many neurodegenerative diseases. Moreover, we summarize different signaling pathways that can be utilized to renovate the 26S proteasome functional capacity, alongside currently known druggable targets in this circuit and various classes of proteasome activators.
Collapse
Affiliation(s)
- Mahmoud A Desouky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Doaa A Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt.
| |
Collapse
|
8
|
Sin YY, Giblin A, Judina A, Rujirachaivej P, Corral LG, Glennon E, Tai ZX, Feng T, Torres E, Zorn A, Gorelik J, Kyurkchieva E, Yenchitsomanus PT, Swindlehurst C, Chan K, Stirling D, Baillie GS. Targeted protein degradation of PDE4 shortforms by a novel proteolysis targeting chimera. FEBS J 2024. [PMID: 39673076 DOI: 10.1111/febs.17359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/17/2024] [Accepted: 10/07/2024] [Indexed: 12/15/2024]
Abstract
Cyclic AMP (cAMP) has a crucial role in many vital cellular processes and there has been much effort expended in the discovery of inhibitors against the enzyme superfamily that degrades this second messenger, namely phosphodiesterases (PDEs). The journey of competitive PDE inhibitors to the clinic has been hampered by side effects profiles that have resulted from a lack of selectivity for subfamilies and individual isoforms because of high conservation of catalytic site sequences and structures. Here we introduce a proteolysis targeting chimera (PROTAC) that can specifically target a small subset of isoforms from the PDE4 family to send the enzyme for degradation at the proteasome by recruiting a ubiquitin E3 ligase into proximity with the PDE. We constructed our PDE4 PROTAC (KTX207) using a previously characterized PDE4 inhibitor, and we show that evolution of the compound into a PROTAC improves selectivity, potency and enables a long-lasting effect even after the compound is removed from cells after a short treatment duration. Functionally, KTX207 is more effective at increasing cAMP, is 100 times more anti-inflammatory, and is significantly better at reducing the growth in cancer cell models than the PDE4 inhibitor alone. Our study highlights the advantages of targeted degradation versus active-site occupancy for PDE4 inhibition and discusses the potential of this novel pharmacological approach to improve the safety profile of PDE4 inhibition in the future.
Collapse
Affiliation(s)
- Yuan Yan Sin
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Aoife Giblin
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Aleksandra Judina
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, UK
| | - Punchita Rujirachaivej
- Graduate Program in Clinical Pathology, Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Eliza Glennon
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Zhi Xian Tai
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Tian Feng
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | | | - Alina Zorn
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Julia Gorelik
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, UK
| | - Elka Kyurkchieva
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Pa Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT) and Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Kyle Chan
- Katalytic Therapeutics, San Diego, CA, USA
| | | | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| |
Collapse
|
9
|
Tenchov R, Sasso JM, Zhou QA. Alzheimer's Disease: Exploring the Landscape of Cognitive Decline. ACS Chem Neurosci 2024; 15:3800-3827. [PMID: 39392435 PMCID: PMC11587518 DOI: 10.1021/acschemneuro.4c00339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. The pathology of AD is marked by the accumulation of amyloid beta plaques and tau protein tangles in the brain, along with neuroinflammation and synaptic dysfunction. Genetic factors, such as mutations in APP, PSEN1, and PSEN2 genes, as well as the APOE ε4 allele, contribute to increased risk of acquiring AD. Currently available treatments provide symptomatic relief but do not halt disease progression. Research efforts are focused on developing disease-modifying therapies that target the underlying pathological mechanisms of AD. Advances in identification and validation of reliable biomarkers for AD hold great promise for enhancing early diagnosis, monitoring disease progression, and assessing treatment response in clinical practice in effort to alleviate the burden of this devastating disease. In this paper, we analyze data from the CAS Content Collection to summarize the research progress in Alzheimer's disease. We examine the publication landscape in effort to provide insights into current knowledge advances and developments. We also review the most discussed and emerging concepts and assess the strategies to combat the disease. We explore the genetic risk factors, pharmacological targets, and comorbid diseases. Finally, we inspect clinical applications of products against AD with their development pipelines and efforts for drug repurposing. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding AD, to outline challenges, and to evaluate growth opportunities to further efforts in combating the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | | |
Collapse
|
10
|
Armstrong P, Güngör H, Anongjanya P, Tweedy C, Parkin E, Johnston J, Carr IM, Dawson N, Clapcote SJ. Protective effect of PDE4B subtype-specific inhibition in an App knock-in mouse model for Alzheimer's disease. Neuropsychopharmacology 2024; 49:1559-1568. [PMID: 38521860 PMCID: PMC11319650 DOI: 10.1038/s41386-024-01852-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 02/24/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
Meta-analysis of genome-wide association study data has implicated PDE4B in the pathogenesis of Alzheimer's disease (AD), the leading cause of senile dementia. PDE4B encodes one of four subtypes of cyclic adenosine monophosphate (cAMP)-specific phosphodiesterase-4 (PDE4A-D). To interrogate the involvement of PDE4B in the manifestation of AD-related phenotypes, the effects of a hypomorphic mutation (Pde4bY358C) that decreases PDE4B's cAMP hydrolytic activity were evaluated in the AppNL-G-F knock-in mouse model of AD using the Barnes maze test of spatial memory, 14C-2-deoxyglucose autoradiography, thioflavin-S staining of β-amyloid (Aβ) plaques, and inflammatory marker assay and transcriptomic analysis (RNA sequencing) of cerebral cortical tissue. At 12 months of age, AppNL-G-F mice exhibited spatial memory and brain metabolism deficits, which were prevented by the hypomorphic PDE4B in AppNL-G-F/Pde4bY358C mice, without a decrease in Aβ plaque burden. RNA sequencing revealed that, among the 531 transcripts differentially expressed in AppNL-G-F versus wild-type mice, only 13 transcripts from four genes - Ide, Btaf1, Padi2, and C1qb - were differentially expressed in AppNL-G-F/Pde4bY358C versus AppNL-G-F mice, identifying their potential involvement in the protective effect of hypomorphic PDE4B. Our data demonstrate that spatial memory and cerebral glucose metabolism deficits exhibited by 12-month-old AppNL-G-F mice are prevented by targeted inhibition of PDE4B. To our knowledge, this is the first demonstration of a protective effect of PDE4B subtype-specific inhibition in a preclinical model of AD. It thus identifies PDE4B as a key regulator of disease manifestation in the AppNL-G-F model and a promising therapeutic target for AD.
Collapse
Affiliation(s)
- Paul Armstrong
- School of Biomedical Sciences, University of Leeds, LS2 9JT, Leeds, UK
| | - Hüseyin Güngör
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, LA1 4YG, Lancaster, UK
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, Cumhuriyet University, Sivas, 58140, Turkey
| | - Pariya Anongjanya
- School of Biomedical Sciences, University of Leeds, LS2 9JT, Leeds, UK
| | - Clare Tweedy
- School of Biomedical Sciences, University of Leeds, LS2 9JT, Leeds, UK
| | - Edward Parkin
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, LA1 4YG, Lancaster, UK
| | - Jamie Johnston
- School of Biomedical Sciences, University of Leeds, LS2 9JT, Leeds, UK
| | - Ian M Carr
- Leeds Institute of Medical Research, University of Leeds, LS9 7TF, Leeds, UK
| | - Neil Dawson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, LA1 4YG, Lancaster, UK
| | - Steven J Clapcote
- School of Biomedical Sciences, University of Leeds, LS2 9JT, Leeds, UK.
| |
Collapse
|
11
|
Donders Z, Skorupska IJ, Willems E, Mussen F, Broeckhoven JV, Carlier A, Schepers M, Vanmierlo T. Beyond PDE4 inhibition: A comprehensive review on downstream cAMP signaling in the central nervous system. Biomed Pharmacother 2024; 177:117009. [PMID: 38908196 DOI: 10.1016/j.biopha.2024.117009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/27/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is a key second messenger that regulates signal transduction pathways pivotal for numerous biological functions. Intracellular cAMP levels are spatiotemporally regulated by their hydrolyzing enzymes called phosphodiesterases (PDEs). It has been shown that increased cAMP levels in the central nervous system (CNS) promote neuroplasticity, neurotransmission, neuronal survival, and myelination while suppressing neuroinflammation. Thus, elevating cAMP levels through PDE inhibition provides a therapeutic approach for multiple CNS disorders, including multiple sclerosis, stroke, spinal cord injury, amyotrophic lateral sclerosis, traumatic brain injury, and Alzheimer's disease. In particular, inhibition of the cAMP-specific PDE4 subfamily is widely studied because of its high expression in the CNS. So far, the clinical translation of full PDE4 inhibitors has been hampered because of dose-limiting side effects. Hence, focusing on signaling cascades downstream activated upon PDE4 inhibition presents a promising strategy, offering novel and pharmacologically safe targets for treating CNS disorders. Yet, the underlying downstream signaling pathways activated upon PDE(4) inhibition remain partially elusive. This review provides a comprehensive overview of the existing knowledge regarding downstream mediators of cAMP signaling induced by PDE4 inhibition or cAMP stimulators. Furthermore, we highlight existing gaps and future perspectives that may incentivize additional downstream research concerning PDE(4) inhibition, thereby providing novel therapeutic approaches for CNS disorders.
Collapse
Affiliation(s)
- Zoë Donders
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Iga Joanna Skorupska
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Emily Willems
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Femke Mussen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht 6629ER, the Netherlands
| | - Melissa Schepers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Tim Vanmierlo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht 6229ER, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt 3500, Belgium; University MS Centre (UMSC) Hasselt - Pelt, Belgium.
| |
Collapse
|
12
|
Lusardi M, Rapetti F, Spallarossa A, Brullo C. PDE4D: A Multipurpose Pharmacological Target. Int J Mol Sci 2024; 25:8052. [PMID: 39125619 PMCID: PMC11311937 DOI: 10.3390/ijms25158052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Phosphodiesterase 4 (PDE4) enzymes catalyze cyclic adenosine monophosphate (cAMP) hydrolysis and are involved in a variety of physiological processes, including brain function, monocyte and macrophage activation, and neutrophil infiltration. Among different PDE4 isoforms, Phosphodiesterases 4D (PDE4Ds) play a fundamental role in cognitive, learning and memory consolidation processes and cancer development. Selective PDE4D inhibitors (PDE4Dis) could represent an innovative and valid therapeutic strategy for the treatment of various neurodegenerative diseases, such as Alzheimer's, Parkinson's, Huntington's, and Lou Gehrig's diseases, but also for stroke, traumatic brain and spinal cord injury, mild cognitive impairment, and all demyelinating diseases such as multiple sclerosis. In addition, small molecules able to block PDE4D isoforms have been recently studied for the treatment of specific cancer types, particularly hepatocellular carcinoma and breast cancer. This review overviews the PDE4DIsso far identified and provides useful information, from a medicinal chemistry point of view, for the development of a novel series of compounds with improved pharmacological properties.
Collapse
Affiliation(s)
- Matteo Lusardi
- Department of Pharmacy (DIFAR), University of Genoa, Viale Benedetto XV 3, 16132 Genova, Italy; (F.R.); (A.S.)
| | | | | | - Chiara Brullo
- Department of Pharmacy (DIFAR), University of Genoa, Viale Benedetto XV 3, 16132 Genova, Italy; (F.R.); (A.S.)
| |
Collapse
|
13
|
Schepers M, Hendrix S, Mussen F, van Breedam E, Ponsaerts P, Lemmens S, Hellings N, Ricciarelli R, Fedele E, Bruno O, Brullo C, Prickaerts J, Van Broeckhoven J, Vanmierlo T. Amelioration of functional and histopathological consequences after spinal cord injury through phosphodiesterase 4D (PDE4D) inhibition. Neurotherapeutics 2024; 21:e00372. [PMID: 38760316 PMCID: PMC11284540 DOI: 10.1016/j.neurot.2024.e00372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/15/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
Spinal cord injury (SCI) is a life-changing event that severely impacts the patient's quality of life. Modulating neuroinflammation, which exacerbates the primary injury, and stimulating neuro-regenerative repair mechanisms are key strategies to improve functional recovery. Cyclic adenosine monophosphate (cAMP) is a second messenger crucially involved in both processes. Following SCI, intracellular levels of cAMP are known to decrease over time. Therefore, preventing cAMP degradation represents a promising strategy to suppress inflammation while stimulating regeneration. Intracellular cAMP levels are controlled by its hydrolyzing enzymes phosphodiesterases (PDEs). The PDE4 family is most abundantly expressed in the central nervous system (CNS) and its inhibition has been shown to be therapeutically relevant for managing SCI pathology. Unfortunately, the use of full PDE4 inhibitors at therapeutic doses is associated with severe emetic side effects, hampering their translation toward clinical applications. Therefore, in this study, we evaluated the effect of inhibiting specific PDE4 subtypes (PDE4B and PDE4D) on inflammatory and regenerative processes following SCI, as inhibitors selective for these subtypes have been demonstrated to be well-tolerated. We reveal that administration of the PDE4D inhibitor Gebr32a, even when starting 2 dpi, but not the PDE4B inhibitor A33, improved functional as well as histopathological outcomes after SCI, comparable to results obtained with the full PDE4 inhibitor roflumilast. Furthermore, using a luminescent human iPSC-derived neurospheroid model, we show that PDE4D inhibition stabilizes neural viability by preventing apoptosis and stimulating neuronal differentiation. These findings strongly suggest that specific PDE4D inhibition offers a novel therapeutic approach for SCI.
Collapse
Affiliation(s)
- Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Sven Hendrix
- Institute for Translational Medicine, Medical School Hamburg, 20457 Hamburg, Germany
| | - Femke Mussen
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Elise van Breedam
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610 Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610 Wilrijk, Belgium
| | - Stefanie Lemmens
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Niels Hellings
- University MS Centre (UMSC) Hasselt - Pelt, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Roberta Ricciarelli
- IRCCS Ospedale Policlinico San Martino, 16100 Genoa, Italy; Department of Experimental Medicine, Section of General Pathology, University of Genova, 16100 Genoa, Italy
| | - Ernesto Fedele
- IRCCS Ospedale Policlinico San Martino, 16100 Genoa, Italy; Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, 16100 Genoa, Italy
| | - Olga Bruno
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, 16100 Genoa, Italy
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, 16100 Genoa, Italy
| | - Jos Prickaerts
- Peitho Translational, 6229ER Maastricht, the Netherlands
| | - Jana Van Broeckhoven
- University MS Centre (UMSC) Hasselt - Pelt, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands; University MS Centre (UMSC) Hasselt - Pelt, Belgium.
| |
Collapse
|
14
|
Sin YY, Cameron RT, Schepers M, MacLeod R, Wright TA, Paes D, van den Hove D, Willems E, Vanmierlo T, Prickaerts J, Blair CM, Baillie GS. Beta-amyloid interacts with and activates the long-form phosphodiesterase PDE4D5 in neuronal cells to reduce cAMP availability. FEBS Lett 2024; 598:1591-1604. [PMID: 38724485 DOI: 10.1002/1873-3468.14902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 07/09/2024]
Abstract
Inhibition of the cyclic-AMP degrading enzyme phosphodiesterase type 4 (PDE4) in the brains of animal models is protective in Alzheimer's disease (AD). We show for the first time that enzymes from the subfamily PDE4D not only colocalize with beta-amyloid (Aβ) plaques in a mouse model of AD but that Aβ directly associates with the catalytic machinery of the enzyme. Peptide mapping suggests that PDE4D is the preferential PDE4 subfamily for Aβ as it possesses a unique binding site. Intriguingly, exogenous addition of Aβ to cells overexpressing the PDE4D5 longform caused PDE4 activation and a decrease in cAMP. We suggest a novel mechanism where PDE4 longforms can be activated by Aβ, resulting in the attenuation of cAMP signalling to promote loss of cognitive function in AD.
Collapse
Affiliation(s)
- Yuan Yan Sin
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Ryan T Cameron
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | - Ruth MacLeod
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Tom A Wright
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Dean Paes
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | - Daniel van den Hove
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | - Emily Willems
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | - Connor M Blair
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| |
Collapse
|
15
|
Rombaut B, Schepers M, Tiane A, Mussen F, Koole L, Kessels S, Trippaers C, Jacobs R, Wouters K, Willems E, van Veggel L, Koulousakis P, Deluyker D, Bito V, Prickaerts J, Wens I, Brône B, van den Hove DLA, Vanmierlo T. Early Inhibition of Phosphodiesterase 4B (PDE4B) Instills Cognitive Resilience in APPswe/PS1dE9 Mice. Cells 2024; 13:1000. [PMID: 38920631 PMCID: PMC11201979 DOI: 10.3390/cells13121000] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/15/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
Microglia activity can drive excessive synaptic loss during the prodromal phase of Alzheimer's disease (AD) and is associated with lowered cyclic adenosine monophosphate (cAMP) due to cAMP phosphodiesterase 4B (PDE4B). This study aimed to investigate whether long-term inhibition of PDE4B by A33 (3 mg/kg/day) can prevent synapse loss and its associated cognitive decline in APPswe/PS1dE9 mice. This model is characterized by a chimeric mouse/human APP with the Swedish mutation and human PSEN1 lacking exon 9 (dE9), both under the control of the mouse prion protein promoter. The effects on cognitive function of prolonged A33 treatment from 20 days to 4 months of age, was assessed at 7-8 months. PDE4B inhibition significantly improved both the working and spatial memory of APPswe/PSdE9 mice after treatment ended. At the cellular level, in vitro inhibition of PDE4B induced microglial filopodia formation, suggesting that regulation of PDE4B activity can counteract microglia activation. Further research is needed to investigate if this could prevent microglia from adopting their 'disease-associated microglia (DAM)' phenotype in vivo. These findings support the possibility that PDE4B is a potential target in combating AD pathology and that early intervention using A33 may be a promising treatment strategy for AD.
Collapse
Affiliation(s)
- Ben Rombaut
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| | - Assia Tiane
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| | - Femke Mussen
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Lisa Koole
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Sofie Kessels
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
| | - Chloë Trippaers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Ruben Jacobs
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
| | - Kristiaan Wouters
- Department of Internal Medicine, Maastricht University Medical Center+ (MUMC+), 6229 ER Maastricht, The Netherlands;
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Emily Willems
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Lieve van Veggel
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| | - Philippos Koulousakis
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Dorien Deluyker
- UHasselt, Cardio & Organ Systems (COST), BIOMED, Agoralaan, 3590 Diepenbeek, Belgium; (D.D.); (V.B.)
| | - Virginie Bito
- UHasselt, Cardio & Organ Systems (COST), BIOMED, Agoralaan, 3590 Diepenbeek, Belgium; (D.D.); (V.B.)
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
| | - Inez Wens
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| | - Bert Brône
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
| | - Daniel L. A. van den Hove
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, 97070 Wuerzburg, Germany
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; (B.R.); (M.S.); (A.T.); (F.M.); (L.K.); (S.K.); (C.T.); (R.J.); (E.W.); (L.v.V.); (P.K.); (I.W.); (B.B.)
- Department Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNs), Division Translational Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.P.); (D.L.A.v.d.H.)
- University MS Center (UMSC) Hasselt, 3900 Pelt, Belgium
| |
Collapse
|
16
|
Li Q, Liao Q, Qi S, Huang H, He S, Lyu W, Liang J, Qin H, Cheng Z, Yu F, Dong X, Wang Z, Han L, Han Y. Opportunities and perspectives of small molecular phosphodiesterase inhibitors in neurodegenerative diseases. Eur J Med Chem 2024; 271:116386. [PMID: 38614063 DOI: 10.1016/j.ejmech.2024.116386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 04/15/2024]
Abstract
Phosphodiesterase (PDE) is a superfamily of enzymes that are responsible for the hydrolysis of two second messengers: cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). PDE inhibition promotes the gene transcription by activating cAMP-response element binding protein (CREB), initiating gene transcription of brain-derived neurotrophic factor (BDNF). The procedure exerts neuroprotective profile, and motor and cognitive improving efficacy. From this point of view, PDE inhibition will provide a promising therapeutic strategy for treating neurodegenerative disorders. Herein, we summarized the PDE inhibitors that have entered the clinical trials or been discovered in recent five years. Well-designed clinical or preclinical investigations have confirmed the effectiveness of PDE inhibitors, such as decreasing Aβ oligomerization and tau phosphorylation, alleviating neuro-inflammation and oxidative stress, modulating neuronal plasticity and improving long-term cognitive impairment.
Collapse
Affiliation(s)
- Qi Li
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| | - Qinghong Liao
- Shandong Kangqiao Biotechnology Co., Ltd, Qingdao, 266033, Shandong, PR China
| | - Shulei Qi
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - He Huang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Siyu He
- Guizhou Province Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, 550004, Guizhou, PR China
| | - Weiping Lyu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Jinxin Liang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Huan Qin
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Zimeng Cheng
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Fan Yu
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Xue Dong
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Ziming Wang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China; School of Pharmacy, Binzhou Medical University, Yantai, 256699, Shandong, PR China
| | - Lingfei Han
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, PR China
| | - Yantao Han
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| |
Collapse
|
17
|
Jino K, Miyamoto K, Kanbara T, Unemura C, Horiguchi N, Ago Y. Allosteric inhibition of phosphodiesterase 4D induces biphasic memory-enhancing effects associated with learning-activated signaling pathways. Psychopharmacology (Berl) 2024; 241:805-816. [PMID: 38114603 DOI: 10.1007/s00213-023-06510-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023]
Abstract
RATIONALE Phosphodiesterase 4D negative allosteric modulators (PDE4D NAMs) enhance memory and cognitive function in animal models without emetic-like side effects. However, the relationship between increased cyclic adenosine monophosphate (cAMP) signaling and the effects of PDE4D NAM remains elusive. OBJECTIVE To investigate the roles of hippocampal cAMP metabolism and synaptic activation in the effects of D159687, a PDE4D NAM, under baseline and learning-stimulated conditions. RESULTS At 3 mg/kg, D159687 enhanced memory formation and consolidation in contextual fear conditioning; however, neither lower (0.3 mg/kg) nor higher (30 mg/kg) doses induced memory-enhancing effects. A biphasic (bell-shaped) dose-response effect was also observed in a scopolamine-induced model of amnesia in the Y-maze, whereas D159687 dose-dependently caused an emetic-like effect in the xylazine/ketamine anesthesia test. At 3 mg/kg, D159687 increased cAMP levels in the hippocampal CA1 region after conditioning in the fear conditioning test, but not in the home-cage or conditioning cage (i.e., context only). By contrast, 30 mg/kg of D159687 increased hippocampal cAMP levels under all conditions. Although both 3 and 30 mg/kg of D159687 upregulated learning-induced Fos expression in the hippocampal CA1 30 min after conditioning, 3 mg/kg, but not 30 mg/kg, of D159687 induced phosphorylation of synaptic plasticity-related proteins such as cAMP-responsive element-binding protein, synaptosomal-associated protein 25 kDa, and the N-methyl-D-aspartate receptor subunit NR2A. CONCLUSIONS Our findings suggest that learning-stimulated conditions can alter the effects of a PDE4D NAM on hippocampal cAMP levels and imply that a PDE4D NAM exerts biphasic memory-enhancing effects associated with synaptic plasticity-related signaling activation.
Collapse
Affiliation(s)
- Kohei Jino
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Keisuke Miyamoto
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
| | - Tomoe Kanbara
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
| | - Chie Unemura
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan
| | - Naotaka Horiguchi
- Laboratory for Drug Discovery and Disease Research, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka-shi, Osaka, 561-0825, Japan.
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan.
| |
Collapse
|
18
|
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: 0.5] [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
|
19
|
Martinez JM, Shen A, Xu B, Jovanovic A, de Chabot J, Zhang J, Xiang YK. Arrestin-dependent nuclear export of phosphodiesterase 4D promotes GPCR-induced nuclear cAMP signaling required for learning and memory. Sci Signal 2023; 16:eade3380. [PMID: 36976866 PMCID: PMC10404024 DOI: 10.1126/scisignal.ade3380] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023]
Abstract
G protein-coupled receptors (GPCRs) promote the expression of immediate early genes required for learning and memory. Here, we showed that β2-adrenergic receptor (β2AR) stimulation induced the nuclear export of phosphodiesterase 4D5 (PDE4D5), an enzyme that degrades the second messenger cAMP, to enable memory consolidation. We demonstrated that the endocytosis of β2AR phosphorylated by GPCR kinases (GRKs) mediated arrestin3-dependent nuclear export of PDE4D5, which was critical for promoting nuclear cAMP signaling and gene expression in hippocampal neurons for memory consolidation. Inhibition of the arrestin3-PDE4D5 association prevented β2AR-induced nuclear cAMP signaling without affecting receptor endocytosis. Direct PDE4 inhibition rescued β2AR-induced nuclear cAMP signaling and ameliorated memory deficits in mice expressing a form of the β2AR that could not be phosphorylated by GRKs. These data reveal how β2AR phosphorylated by endosomal GRK promotes the nuclear export of PDE4D5, leading to nuclear cAMP signaling, changes in gene expression, and memory consolidation. This study also highlights the translocation of PDEs as a mechanism to promote cAMP signaling in specific subcellular locations downstream of GPCR activation.
Collapse
Affiliation(s)
- Joseph M. Martinez
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Ao Shen
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
- School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Bing Xu
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
- VA Northern California Health Care System, Mather, CA, 95655, USA
| | - Aleksandra Jovanovic
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Josephine de Chabot
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
| | - Jin Zhang
- Department of Pharmacology, University of California at San Diego, San Diego, CA, 92093, USA
| | - Yang K. Xiang
- Department of Pharmacology, University of California at Davis, Davis, CA, 95616, USA
- VA Northern California Health Care System, Mather, CA, 95655, USA
| |
Collapse
|
20
|
Schepers M, Paes D, Tiane A, Rombaut B, Piccart E, van Veggel L, Gervois P, Wolfs E, Lambrichts I, Brullo C, Bruno O, Fedele E, Ricciarelli R, Ffrench-Constant C, Bechler ME, van Schaik P, Baron W, Lefevere E, Wasner K, Grünewald A, Verfaillie C, Baeten P, Broux B, Wieringa P, Hellings N, Prickaerts J, Vanmierlo T. Selective PDE4 subtype inhibition provides new opportunities to intervene in neuroinflammatory versus myelin damaging hallmarks of multiple sclerosis. Brain Behav Immun 2023; 109:1-22. [PMID: 36584795 DOI: 10.1016/j.bbi.2022.12.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/17/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) characterized by focal inflammatory lesions and prominent demyelination. Even though the currently available therapies are effective in treating the initial stages of disease, they are unable to halt or reverse disease progression into the chronic progressive stage. Thus far, no repair-inducing treatments are available for progressive MS patients. Hence, there is an urgent need for the development of new therapeutic strategies either targeting the destructive immunological demyelination or boosting endogenous repair mechanisms. Using in vitro, ex vivo, and in vivo models, we demonstrate that selective inhibition of phosphodiesterase 4 (PDE4), a family of enzymes that hydrolyzes and inactivates cyclic adenosine monophosphate (cAMP), reduces inflammation and promotes myelin repair. More specifically, we segregated the myelination-promoting and anti-inflammatory effects into a PDE4D- and PDE4B-dependent process respectively. We show that inhibition of PDE4D boosts oligodendrocyte progenitor cells (OPC) differentiation and enhances (re)myelination of both murine OPCs and human iPSC-derived OPCs. In addition, PDE4D inhibition promotes in vivo remyelination in the cuprizone model, which is accompanied by improved spatial memory and reduced visual evoked potential latency times. We further identified that PDE4B-specific inhibition exerts anti-inflammatory effects since it lowers in vitro monocytic nitric oxide (NO) production and improves in vivo neurological scores during the early phase of experimental autoimmune encephalomyelitis (EAE). In contrast to the pan PDE4 inhibitor roflumilast, the therapeutic dose of both the PDE4B-specific inhibitor A33 and the PDE4D-specific inhibitor Gebr32a did not trigger emesis-like side effects in rodents. Finally, we report distinct PDE4D isoform expression patterns in human area postrema neurons and human oligodendroglia lineage cells. Using the CRISPR-Cas9 system, we confirmed that pde4d1/2 and pde4d6 are the key targets to induce OPC differentiation. Collectively, these data demonstrate that gene specific PDE4 inhibitors have potential as novel therapeutic agents for targeting the distinct disease processes of MS.
Collapse
Affiliation(s)
- Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands; University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Dean Paes
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Assia Tiane
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands; University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Ben Rombaut
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Elisabeth Piccart
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Lieve van Veggel
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands; University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Pascal Gervois
- Department of Cardio and Organ Systems, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Esther Wolfs
- Department of Cardio and Organ Systems, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Ivo Lambrichts
- Department of Cardio and Organ Systems, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, Genova, Italy
| | - Olga Bruno
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, Genova, Italy
| | - Ernesto Fedele
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Roberta Ricciarelli
- IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Experimental Medicine, Section of General Pathology, University of Genova, Genova, Italy
| | - Charles Ffrench-Constant
- MRC Centre for Regenerative Medicine and MS Society Edinburgh Centre, Edinburgh bioQuarter, University of Edinburgh, Edinburgh, UK
| | - Marie E Bechler
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Pauline van Schaik
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Wia Baron
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Evy Lefevere
- Rewind Therapeutics NV, Gaston Geenslaan 2, B-3001, Leuven, Belgium
| | - Kobi Wasner
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Catherine Verfaillie
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Belgium
| | - Paulien Baeten
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Bieke Broux
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Paul Wieringa
- MERLN Institute for Technology-Inspired Regenerative Medicine, Complex Tissue Regeneration department, Maastricht University, Maastricht, the Netherlands
| | - Niels Hellings
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jos Prickaerts
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands; University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium.
| |
Collapse
|
21
|
Targeting phosphodiesterase 4 as a therapeutic strategy for cognitive improvement. Bioorg Chem 2022; 130:106278. [DOI: 10.1016/j.bioorg.2022.106278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/22/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022]
|
22
|
Liu P, Wang J, Peng S, Zhang D, Zhuang L, Liu C, Zhang Y, Shi X. Suppression of phosphodiesterase IV enzyme by roflumilast ameliorates cognitive dysfunction in aged rats after sevoflurane anaesthesia via PKA-CREB and MEK/ERK pathways. Eur J Neurosci 2022; 56:4317-4332. [PMID: 35767003 DOI: 10.1111/ejn.15751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022]
Abstract
Postoperative cognitive dysfunction (POCD) is a prevalent disorder after anaesthesia in the elderly patients. Roflumilast (RF), a phosphodiesterase 4 (PDE-4) inhibitor, could improve cognition with no side effects. Here, we sought to explore the efficacy of RF in the improvement of cognitive dysfunction caused by sevoflurane (Sev). Sprague-Dawley rats were anaesthetized, and the hippocampal neurons were treated with Sev to develop in vivo and in vitro POCD models, followed by RF administration. The mechanism of the PKA-CREB and MEK/ERK pathways in the pathogenesis of POCD was explored. Sev impaired the cognitive functions of rats, significantly reduced cyclic adenosine monophosphate (cAMP) concentrations and blocked the PKA-CREB and MEK/ERK pathways. Moreover, the Sev-treated rats and neurons exhibited enhanced apoptosis and reactive oxygen species (ROS). After treatment with RF, rats had better learning and memory function, and the activity of neurons in hippocampus and cortex was improved. Loss-of-function assay indicated that PKA-CREB and MEK/ERK signalling impairment reduced cAMP levels and promoted apoptosis and ROS in rat hippocampus and neurons. Generally, RF promotes neuronal activity in rats after Sev treatment by maintaining cAMP levels and sustaining the activation of PKA-CREB and MEK/ERK pathways. This might offer novel sights for POCD therapy.
Collapse
Affiliation(s)
- Peirong Liu
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, Shanghai, China
| | - Juan Wang
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, Shanghai, China
| | - Sheng Peng
- Department of Anesthesiology, Longhua Hospital Shanghai University of TCM, Shanghai, China
| | - Dan Zhang
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, Shanghai, China
| | - Lin Zhuang
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, Shanghai, China
| | - Chunliang Liu
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, Shanghai, China
| | - Yu Zhang
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, Shanghai, China
| | - Xiaowei Shi
- Department of Anesthesiology, Seventh People's Hospital of Shanghai University of TCM, Shanghai, China
| |
Collapse
|
23
|
Zhou F, Huang Y, Liu L, Song Z, Hou KQ, Yang Y, Luo HB, Huang YY, Xiong XF. Structure-based optimization of Toddacoumalone as highly potent and selective PDE4 inhibitors with anti-inflammatory effects. Biochem Pharmacol 2022; 202:115123. [PMID: 35688178 DOI: 10.1016/j.bcp.2022.115123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 11/29/2022]
Abstract
Phosphodiesterase-4 (PDE4) is an important drug target for inflammatory diseases. Previously, we identified a series of novel PDE4 inhibitors derived from the natural Toddacoumalone, among which the hit compound 2 with a naphthyridine scaffold showed moderate potency with the IC50 value of 400 nM. Based on the co-crystal structure of PDE4D-2, further structural optimizations and structure-activity relationship studies led to a highly potent PDE4 inhibitor 23a with the IC50 value of 0.25 nM and excellent selectivity profiles over other PDEs (>4000-fold). The co-crystal structure of PDE4D-23a elucidated that 23a has strong interactions with the M and Q pocket of PDE4D. Importantly, compound 23a significantly inhibits the release of inflammatory cytokines TNF-α and IL-6 in lipopolysaccharide-stimulated RAW264.7 cells. Thus, compound 23a with a naphthyridine scaffold is a promising PDE4 inhibitor for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Feng Zhou
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, PR China
| | - Yue Huang
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, PR China
| | - Lu Liu
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, PR China
| | - Zhendong Song
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, PR China
| | - Ke-Qiang Hou
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, PR China
| | - Yifan Yang
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, PR China
| | - Hai-Bin Luo
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, PR China; Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, 570228 Haikou, PR China
| | - Yi-You Huang
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, PR China; Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, 570228 Haikou, PR China.
| | - Xiao-Feng Xiong
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006 Guangzhou, PR China.
| |
Collapse
|
24
|
Gao F, Yang S, Wang J, Zhu G. cAMP-PKA cascade: An outdated topic for depression? Biomed Pharmacother 2022; 150:113030. [PMID: 35486973 DOI: 10.1016/j.biopha.2022.113030] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/02/2022] Open
Abstract
Depression is a common neuropsychiatric disorder characterized by persistent depressed mood and causes serious socioeconomic burden worldwide. Hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis, deficiency of monoamine transmitters, neuroinflammation and abnormalities of the gut flora are strongly associated with the onset of depression. The cyclic AMP (cAMP)/protein kinase A (PKA) cascade, a major cross-species cellular signaling pathway, is supposed as important player and regulator of depression onset by controlling synaptic plasticity, cytokinesis, transcriptional regulation and HPA axis. In the central nervous system, the cAMP-PKA cascade can dynamically shape neural circuits by enhancing synaptic plasticity, and affect K+ channels by phosphorylating Kir4.1, thereby regulating neuronal excitation. The reduction of cAMP-PKA cascade affects neuronal excitation as well as synaptic plasticity, ultimately leading to pathological outcome of depression, while activation of cAMP-PKA cascade would provide a rapid antidepressant effect. In this review, we proposed to reconsider the function of cAMP-PKA cascade, especially in the rapid antidepressant effect. Local activation or indirect activation of PKA through adjusting anchor proteins would provide new idea for acute treatment of depression.
Collapse
Affiliation(s)
- Feng Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Shaojie Yang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Juan Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
25
|
Xi M, Sun T, Chai S, Xie M, Chen S, Deng L, Du K, Shen R, Sun H. Therapeutic potential of phosphodiesterase inhibitors for cognitive amelioration in Alzheimer's disease. Eur J Med Chem 2022; 232:114170. [DOI: 10.1016/j.ejmech.2022.114170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 02/07/2023]
|
26
|
Liu Z, Liu M, Cao Z, Qiu P, Song G. Phosphodiesterase‑4 inhibitors: a review of current developments (2013-2021). Expert Opin Ther Pat 2022; 32:261-278. [PMID: 34986723 DOI: 10.1080/13543776.2022.2026328] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cyclic nucleotide phosphodiesterase 4 (PDE4) is responsible for the hydrolysis of cAMP, which has become an attractive therapeutic target for lung, skin, and severe neurological diseases. Here, we review the current status of development of PDE4 inhibitors since 2013 and discuss the applicability of novel medicinal-chemistry strategies for identifying more efficient and safer inhibitors. AREAS COVERED This review summarizes the clinical development of PDE4 inhibitors from 2013 to 2021, focused on their pharmacophores, the strategies to reduce the side effects of PDE4 inhibitors and the development of subfamily selective PDE4 inhibitors. EXPERT OPINION To date, great efforts have been made in the development of PDE4 inhibitors, and researchers have established a comprehensive preclinical database and collected some promising data from clinical trials. Although four small-molecule PDE4 inhibitors have been approved by FDA for the treatment of human diseases up to now, further development of other reported PDE4 inhibitors with strong potency has been hampered due to the occurrence of severe side effects. There are currently three main strategies for overcoming the dose limitation and systemic side effects, which provide new opportunities for the clinical development of new PDE4 inhibitors.
Collapse
Affiliation(s)
- Zhihao Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, China
| | - Mingjian Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, China
| | - Zhenqing Cao
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, China
| | - Pengsen Qiu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, China
| | - Gaopeng Song
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, China
| |
Collapse
|
27
|
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: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
28
|
Ponsaerts L, Alders L, Schepers M, de Oliveira RMW, Prickaerts J, Vanmierlo T, Bronckaers A. Neuroinflammation in Ischemic Stroke: Inhibition of cAMP-Specific Phosphodiesterases (PDEs) to the Rescue. Biomedicines 2021; 9:703. [PMID: 34206420 PMCID: PMC8301462 DOI: 10.3390/biomedicines9070703] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/27/2022] Open
Abstract
Ischemic stroke is caused by a thromboembolic occlusion of a major cerebral artery, with the impaired blood flow triggering neuroinflammation and subsequent neuronal damage. Both the innate immune system (e.g., neutrophils, monocytes/macrophages) in the acute ischemic stroke phase and the adaptive immune system (e.g., T cells, B cells) in the chronic phase contribute to this neuroinflammatory process. Considering that the available therapeutic strategies are insufficiently successful, there is an urgent need for novel treatment options. It has been shown that increasing cAMP levels lowers neuroinflammation. By inhibiting cAMP-specific phosphodiesterases (PDEs), i.e., PDE4, 7, and 8, neuroinflammation can be tempered through elevating cAMP levels and, thereby, this can induce an improved functional recovery. This review discusses recent preclinical findings, clinical implications, and future perspectives of cAMP-specific PDE inhibition as a novel research interest for the treatment of ischemic stroke. In particular, PDE4 inhibition has been extensively studied, and is promising for the treatment of acute neuroinflammation following a stroke, whereas PDE7 and 8 inhibition more target the T cell component. In addition, more targeted PDE4 gene inhibition, or combined PDE4 and PDE7 or 8 inhibition, requires more extensive research.
Collapse
Affiliation(s)
- Laura Ponsaerts
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
| | - Lotte Alders
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
| | - Melissa Schepers
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | | | - Jos Prickaerts
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Tim Vanmierlo
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, European Graduate School of Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Annelies Bronckaers
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; (L.P.); (L.A.); (M.S.)
- European Graduate School of Neuroscience (EURON), Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
29
|
Brullo C, Rapetti F, Abbate S, Prosdocimi T, Torretta A, Semrau M, Massa M, Alfei S, Storici P, Parisini E, Bruno O. Design, synthesis, biological evaluation and structural characterization of novel GEBR library PDE4D inhibitors. Eur J Med Chem 2021; 223:113638. [PMID: 34171658 DOI: 10.1016/j.ejmech.2021.113638] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 11/30/2022]
Abstract
Memory and cognitive functions depend on the cerebral levels of cyclic adenosine monophosphate (cAMP), which are regulated by the phosphodiesterase 4 (PDE4) family of enzymes. Selected rolipram-related PDE4 inhibitors, members of the GEBR library, have been shown to increase hippocampal cAMP levels, providing pro-cognitive benefits with a safe pharmacological profile. In a recent SAR investigation involving a subset of GEBR library compounds, we have demonstrated that, depending on length and flexibility, ligands can either adopt a twisted, an extended or a protruding conformation, the latter allowing the ligand to form stabilizing contacts with the regulatory domain of the enzyme. Here, based on those findings, we describe further chemical modifications of the protruding subset of GEBR library inhibitors and their effects on ligand conformation and potency. In particular, we demonstrate that the insertion of a methyl group in the flexible linker region connecting the catechol portion and the basic end of the molecules enhances the ability of the ligand to interact with both the catalytic and the regulatory domains of the enzyme.
Collapse
Affiliation(s)
- Chiara Brullo
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genova, Viale Benedetto XV 3, 16132, Genova, Italy
| | - Federica Rapetti
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genova, Viale Benedetto XV 3, 16132, Genova, Italy
| | - Sara Abbate
- Center for Nano Science and Technology @ PoliMi, Istituto Italiano di Tecnologia, via Giovanni Pascoli 70/3, 20133, Milano, Italy
| | - Tommaso Prosdocimi
- Center for Nano Science and Technology @ PoliMi, Istituto Italiano di Tecnologia, via Giovanni Pascoli 70/3, 20133, Milano, Italy
| | - Archimede Torretta
- Center for Nano Science and Technology @ PoliMi, Istituto Italiano di Tecnologia, via Giovanni Pascoli 70/3, 20133, Milano, Italy
| | - Marta Semrau
- Elettra Sincrotrone Trieste S.C.p.A., SS 14 - km 163,5 in AREA Science Park, 34149, Trieste, Italy
| | - Matteo Massa
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genova, Viale Benedetto XV 3, 16132, Genova, Italy
| | - Silvana Alfei
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genova, Viale Benedetto XV 3, 16132, Genova, Italy
| | - Paola Storici
- Elettra Sincrotrone Trieste S.C.p.A., SS 14 - km 163,5 in AREA Science Park, 34149, Trieste, Italy
| | - Emilio Parisini
- Center for Nano Science and Technology @ PoliMi, Istituto Italiano di Tecnologia, via Giovanni Pascoli 70/3, 20133, Milano, Italy; Latvian Institute of Organic Synthesis, Aizkraukles 21, LV, 1006, Riga, Latvia.
| | - Olga Bruno
- Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genova, Viale Benedetto XV 3, 16132, Genova, Italy.
| |
Collapse
|
30
|
Furlan V, Bren U. Insight into Inhibitory Mechanism of PDE4D by Dietary Polyphenols Using Molecular Dynamics Simulations and Free Energy Calculations. Biomolecules 2021; 11:479. [PMID: 33806914 PMCID: PMC8004924 DOI: 10.3390/biom11030479] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022] Open
Abstract
Phosphodiesterase 4 (PDE4), mainly present in immune, epithelial, and brain cells, represents a family of key enzymes for the degradation of cyclic adenosine monophosphate (cAMP), which modulates inflammatory response. In recent years, the inhibition of PDE4 has been proven to be an effective therapeutic strategy for the treatment of neurological disorders. PDE4D constitutes a high-interest therapeutic target primarily for the treatment of Alzheimer's disease, as it is highly involved in neuroinflammation, learning ability, and memory dysfunctions. In the present study, a thorough computational investigation consisting of molecular docking, molecular dynamics (MD) simulations, and binding free energy calculations based on the linear response approximation (LRA) method was performed to study dietary polyphenols as potential PDE4D inhibitors. The obtained results revealed that curcumin, 6-gingerol, capsaicin, and resveratrol represent potential PDE4D inhibitors; however, the predicted binding free energies of 6-gingerol, capsaicin, and resveratrol were less negative than in the case of curcumin, which exhibited the highest inhibitory potency in comparison with a positive control rolipram. Our results also revealed that the electrostatic component through hydrogen bonding represents the main driving force for the binding and inhibitory activity of curcumin, 6-gingerol, and resveratrol, while the van der Waals component through shape complementarity plays the most important role in capsaicin's inhibitory activity. All investigated compounds form hydrophobic interactions with residues Gln376 and Asn602 as well as hydrogen bonds with nearby residues Asp438, Met439, and Ser440. The binding mode of the studied natural compounds is consequently very similar; however, it significantly differs from the binding of known PDE4 inhibitors. The uncovered molecular inhibitory mechanisms of four investigated natural polyphenols, curcumin, 6-gingerol, capsaicin, and resveratrol, form the basis for the design of novel PDE4D inhibitors for the treatment of Alzheimer's disease with a potentially wider therapeutic window and fewer adverse side effects.
Collapse
Affiliation(s)
- Veronika Furlan
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia;
| | - Urban Bren
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova 17, SI-2000 Maribor, Slovenia;
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, SI-6000 Koper, Slovenia
| |
Collapse
|
31
|
Gorny N, Kelly MP. Alterations in cyclic nucleotide signaling are implicated in healthy aging and age-related pathologies of the brain. VITAMINS AND HORMONES 2021; 115:265-316. [PMID: 33706951 DOI: 10.1016/bs.vh.2020.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is not only important to consider how hormones may change with age, but also how downstream signaling pathways that couple to hormone receptors may change. Among these hormone-coupled signaling pathways are the 3',5'-cyclic guanosine monophosphate (cGMP) and 3',5'-cyclic adenosine monophosphate (cAMP) intracellular second messenger cascades. Here, we test the hypothesis that dysfunction of cAMP and/or cGMP synthesis, execution, and/or degradation occurs in the brain during healthy and pathological diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Although most studies report lower cyclic nucleotide signaling in the aged brain, with further reductions noted in the context of age-related diseases, there are select examples where cAMP signaling may be elevated in select tissues. Thus, therapeutics would need to target cAMP/cGMP in a tissue-specific manner if efficacy for select symptoms is to be achieved without worsening others.
Collapse
Affiliation(s)
- Nicole Gorny
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Michy P Kelly
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
32
|
Memory Enhancers for Alzheimer's Dementia: Focus on cGMP. Pharmaceuticals (Basel) 2021; 14:ph14010061. [PMID: 33451088 PMCID: PMC7828493 DOI: 10.3390/ph14010061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Cyclic guanosine-3',5'-monophosphate, better known as cyclic-GMP or cGMP, is a classical second messenger involved in a variety of intracellular pathways ultimately controlling different physiological functions. The family of guanylyl cyclases that includes soluble and particulate enzymes, each of which comprises several isoforms with different mechanisms of activation, synthesizes cGMP. cGMP signaling is mainly executed by the activation of protein kinase G and cyclic nucleotide gated channels, whereas it is terminated by its hydrolysis to GMP operated by both specific and dual-substrate phosphodiesterases. In the central nervous system, cGMP has attracted the attention of neuroscientists especially for its key role in the synaptic plasticity phenomenon of long-term potentiation that is instrumental to memory formation and consolidation, thus setting off a "gold rush" for new drugs that could be effective for the treatment of cognitive deficits. In this article, we summarize the state of the art on the neurochemistry of the cGMP system and then review the pre-clinical and clinical evidence on the use of cGMP enhancers in Alzheimer's disease (AD) therapy. Although preclinical data demonstrates the beneficial effects of cGMP on cognitive deficits in AD animal models, the results of the clinical studies carried out to date are not conclusive. More trials with a dose-finding design on selected AD patient's cohorts, possibly investigating also combination therapies, are still needed to evaluate the clinical potential of cGMP enhancers.
Collapse
|
33
|
Leslie SN, Datta D, Christensen KR, van Dyck CH, Arnsten AFT, Nairn AC. Phosphodiesterase PDE4D Is Decreased in Frontal Cortex of Aged Rats and Positively Correlated With Working Memory Performance and Inversely Correlated With PKA Phosphorylation of Tau. Front Aging Neurosci 2020; 12:576723. [PMID: 33192469 PMCID: PMC7655962 DOI: 10.3389/fnagi.2020.576723] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/24/2020] [Indexed: 01/16/2023] Open
Abstract
Age is the largest risk factor for Alzheimer’s disease (AD) and contributes to cognitive impairment in otherwise healthy individuals. Thus, it is critical that we better understand the risk aging presents to vulnerable regions of the brain and carefully design therapeutics to address those effects. In this study we examined age-related changes in cAMP-regulatory protein, phosphodiesterase 4D (PDE4D). Inhibition of PDE4D is currently under investigation as a therapeutic target for AD based on memory-enhancing effects in rodent hippocampus. Therefore, it is important to understand the role of PDE4D in brain regions particularly vulnerable to disease such as the frontal association cortex (FC), where cAMP signaling can impair working memory via opening of potassium channels. We found that PDE4D protein level was decreased in the FC of both moderately and extremely aged rats, and that PDE4D level was correlated with performance on a FC-dependent working memory task. In extremely aged rats, PDE4D was also inversely correlated with levels of phosphorylated tau at serine 214 (S214), a site phosphorylated by protein kinase A. In vitro studies of the PDE4D inhibitor, GEBR-7b, further illustrated that inhibition of PDE4D activity enhanced phosphorylation of tau. pS214-tau phosphorylation is associated with early AD tau pathology, promotes tau dissociation from microtubules and primes subsequent tau hyperphosphorylation at other critical AD-related sites. Age-related loss of PDE4D may thus contribute to the specific vulnerability of the FC to degeneration in AD, and play a critical role in normal cAMP regulation, cautioning against the use of pan-PDE4D inhibitors as therapeutics.
Collapse
Affiliation(s)
- Shannon N Leslie
- Interdepartmental Neuroscience Program, Yale University, School of Medicine, New Haven, CT, United States.,Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States
| | - Dibyadeep Datta
- Department of Neuroscience, Yale University, School of Medicine, New Haven, CT, United States
| | - Kyle R Christensen
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States
| | - Christopher H van Dyck
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States.,Department of Neuroscience, Yale University, School of Medicine, New Haven, CT, United States.,Department of Neurology, Yale University, School of Medicine, New Haven, CT, United States
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University, School of Medicine, New Haven, CT, United States
| | - Angus C Nairn
- Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States
| |
Collapse
|
34
|
Sharma VK, Singh TG, Singh S. Cyclic Nucleotides Signaling and Phosphodiesterase Inhibition: Defying Alzheimer's Disease. Curr Drug Targets 2020; 21:1371-1384. [PMID: 32718286 DOI: 10.2174/1389450121666200727104728] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Defects in brain functions associated with aging and neurodegenerative diseases benefit insignificantly from existing options, suggesting that there is a lack of understanding of pathological mechanisms. Alzheimer's disease (AD) is such a nearly untreatable, allied to age neurological deterioration for which only the symptomatic cure is available and the agents able to mould progression of the disease, is still far away. The altered expression of phosphodiesterases (PDE) and deregulated cyclic nucleotide signaling in AD has provoked a new thought of targeting cyclic nucleotide signaling in AD. Targeting cyclic nucleotides as an intracellular messenger seems to be a viable approach for certain biological processes in the brain and controlling substantial. Whereas, the synthesis, execution, and/or degradation of cyclic nucleotides has been closely linked to cognitive deficits. In relation to cognition, the cyclic nucleotides (cAMP and cGMP) have an imperative execution in different phases of memory, including gene transcription, neurogenesis, neuronal circuitry, synaptic plasticity and neuronal survival, etc. AD is witnessed by impairments of these basic processes underlying cognition, suggesting a crucial role of cAMP/cGMP signaling in AD populations. Phosphodiesterase inhibitors are the exclusive set of enzymes to facilitate hydrolysis and degradation of cAMP and cGMP thereby, maintains their optimum levels initiating it as an interesting target to explore. The present work reviews a neuroprotective and substantial influence of PDE inhibition on physiological status, pathological progression and neurobiological markers of AD in consonance with the intensities of cAMP and cGMP.
Collapse
Affiliation(s)
- Vivek K Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India,Govt. College of Pharmacy, Rohru, District Shimla, Himachal Pradesh-171207, India
| | - Thakur G Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
35
|
Increased isoform-specific phosphodiesterase 4D expression is associated with pathology and cognitive impairment in Alzheimer's disease. Neurobiol Aging 2020; 97:56-64. [PMID: 33157432 DOI: 10.1016/j.neurobiolaging.2020.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/16/2020] [Accepted: 10/04/2020] [Indexed: 02/07/2023]
Abstract
Pharmacological phosphodiesterase 4D (PDE4D) inhibition shows therapeutic potential to restore memory function in Alzheimer's disease (AD), but will likely evoke adverse side effects. As PDE4D encodes multiple isoforms, targeting specific isoforms may improve treatment efficacy and safety. Here, we investigated whether PDE4D isoform expression and PDE4D DNA methylation is affected in AD and whether expression changes are associated with severity of pathology and cognitive impairment. In post-mortem temporal lobe brain material from AD patients (n = 42) and age-matched controls (n = 40), we measured PDE4D isoform expression and PDE4D DNA (hydroxy)methylation using quantitative polymerase chain reaction and Illumina 450k Beadarrays, respectively. Linear regression revealed increased PDE4D1, -D3, -D5, and -D8 expression in AD with concurrent (hydroxy)methylation changes in associated promoter regions. Moreover, increased PDE4D1 and -D3 expression was associated with higherplaque and tau pathology levels, higher Braak stages, and progressed cognitive impairment. Future studies should indicate functional roles of specific PDE4D isoforms and the efficacy and safety of their selective inhibition to restore memory function in AD.
Collapse
|
36
|
Roflumilast: A potential drug for the treatment of cognitive impairment? Neurosci Lett 2020; 736:135281. [DOI: 10.1016/j.neulet.2020.135281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/19/2020] [Accepted: 07/25/2020] [Indexed: 12/16/2022]
|
37
|
Xiang J, Wang X, Gao Y, Li T, Cao R, Yan T, Ma Y, Niu Y, Xue J, Wang B. Phosphodiesterase 4D Gene Modifies the Functional Network of Patients With Mild Cognitive Impairment and Alzheimer's Disease. Front Genet 2020; 11:890. [PMID: 32849849 PMCID: PMC7423997 DOI: 10.3389/fgene.2020.00890] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that is affected by several genetic variants. It has been demonstrated that genetic variants affect brain organization and function. In this study, using whole genome-wide association studies (GWAS), we analyzed the functional magnetic resonance imaging and genetic data from the Alzheimer’s Disease Neuroimaging Initiative dataset (ADNI) dataset and identified genetic variants associated with the topology of the functional brain network http://www.adni-info.org. We found three novel loci (rs2409627, rs9647533, and rs11955845) in an intron of the phosphodiesterase 4D (PDE4D) gene that contribute to abnormalities in the topological organization of the functional network. In particular, compared to the wild-type genotype, the subjects carrying the PDE4D variants had altered network properties, including a significantly reduced clustering coefficient, small-worldness, global and local efficiency, a significantly enhanced path length and a normalized path length. In addition, we found that all global brain network attributes were affected by PDE4D variants to different extents as the disease progressed. Additionally, brain regions with alterations in nodal efficiency due to the variations in PDE4D were predominant in the limbic lobe, temporal lobe and frontal lobes. PDE4D has a great effect on memory consolidation and cognition through long-term potentiation (LTP) effects and/or the promotion of inflammatory effects. PDE4D variants might be a main reasons underlyling for the abnormal topological properties and cognitive impairment. Furthermore, we speculated that PDE4D is a risk factor for neural degenerative diseases and provided important clues for the earlier detection and therapeutic intervention for AD.
Collapse
Affiliation(s)
- Jie Xiang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Xin Wang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Yuan Gao
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Ting Li
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Rui Cao
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Ting Yan
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, China
| | - Yunxiao Ma
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Yan Niu
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Jiayue Xue
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| | - Bin Wang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
| |
Collapse
|
38
|
Dominant-Negative Attenuation of cAMP-Selective Phosphodiesterase PDE4D Action Affects Learning and Behavior. Int J Mol Sci 2020; 21:ijms21165704. [PMID: 32784895 PMCID: PMC7460819 DOI: 10.3390/ijms21165704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 07/26/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
PDE4 cyclic nucleotide phosphodiesterases reduce 3′, 5′ cAMP levels in the CNS and thereby regulate PKA activity and the phosphorylation of CREB, fundamental to depression, cognition, and learning and memory. The PDE4 isoform PDE4D5 interacts with the signaling proteins β-arrestin2 and RACK1, regulators of β2-adrenergic and other signal transduction pathways. Mutations in PDE4D in humans predispose to acrodysostosis, associated with cognitive and behavioral deficits. To target PDE4D5, we developed mice that express a PDE4D5-D556A dominant-negative transgene in the brain. Male transgenic mice demonstrated significant deficits in hippocampus-dependent spatial learning, as assayed in the Morris water maze. In contrast, associative learning, as assayed in a fear conditioning assay, appeared to be unaffected. Male transgenic mice showed augmented activity in prolonged (2 h) open field testing, while female transgenic mice showed reduced activity in the same assay. Transgenic mice showed no demonstrable abnormalities in prepulse inhibition. There was also no detectable difference in anxiety-like behavior, as measured in the elevated plus-maze. These data support the use of a dominant-negative approach to the study of PDE4D5 function in the CNS and specifically in learning and memory.
Collapse
|
39
|
Wang H, Zhang FF, Xu Y, Fu HR, Wang XD, Wang L, Chen W, Xu XY, Gao YF, Zhang JG, Zhang HT. The Phosphodiesterase-4 Inhibitor Roflumilast, a Potential Treatment for the Comorbidity of Memory Loss and Depression in Alzheimer's Disease: A Preclinical Study in APP/PS1 Transgenic Mice. Int J Neuropsychopharmacol 2020; 23:700-711. [PMID: 32645141 PMCID: PMC7727475 DOI: 10.1093/ijnp/pyaa048] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 06/24/2020] [Accepted: 07/07/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Depression is highly related to Alzheimer's disease (AD), yet no effective treatment is available. Phosphodiesterase-4 (PDE4) has been considered a promising target for treatment of AD and depression. Roflumilast, the first PDE4 inhibitor approved for clinical use, improves cognition at doses that do not cause side effects such as emesis. METHODS Here we examined the effects of roflumilast on behavioral dysfunction and the related mechanisms in APPswe/PS1dE9 transgenic mice, a widely used model of AD. Mice at 10 months of age were examined for memory in the novel object recognition and Morris water-maze tests and depression-like behavior in the tail-suspension test and forced swimming test before killing for neurochemical assays. RESULTS In the novel object recognition and Morris water-maze, APPswe/PS1dE9 mice showed significant cognitive declines, which were reversed by roflumilast at 5 and 10 mg/kg orally once per day. In the tail-suspension test and forced swimming test, the AD mice showed prolonged immobility time, which was also reversed by roflumilast. In addition, the staining of hematoxylin-eosin and Nissl showed that roflumilast relieved the neuronal cell injuries, while terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labelling analysis indicated that roflumilast ameliorated cell apoptosis in AD mice. Further, roflumilast reversed the decreased ratio of B-cell lymphoma-2/Bcl-2-associated X protein and the increased expression of PDE4B and PDE4D in the cerebral cortex and hippocampus of AD mice. Finally, roflumilast reversed the decreased levels of cyclic AMP (cAMP) and expression of phosphorylated cAMP response element-binding protein and brain derived neurotrophic factor in AD mice. CONCLUSIONS Together, these results suggest that roflumilast not only improves learning and memory but also attenuates depression-like behavior in AD mice, likely via PDE4B/PDE4D-mediated cAMP/cAMP response element-binding protein/brain derived neurotrophic factor signaling. Roflumilast can be a therapeutic agent for AD, in particular the comorbidity of memory loss and depression.
Collapse
Affiliation(s)
- Hao Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Fang-fang Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Yong Xu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Hua-rong Fu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Xiao-dan Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Lei Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Wei Chen
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Xiao-yan Xu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Yong-feng Gao
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Ji-guo Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine & Psychiatry, the Rockefeller Neuroscience Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia,Correspondence: Han-Ting Zhang, MD, PhD, Department of Neuroscience, the Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV 26506 ()
| |
Collapse
|
40
|
Ribaudo G, Ongaro A, Zagotto G, Memo M, Gianoncelli A. Therapeutic Potential of Phosphodiesterase Inhibitors against Neurodegeneration: The Perspective of the Medicinal Chemist. ACS Chem Neurosci 2020; 11:1726-1739. [PMID: 32401481 PMCID: PMC8007108 DOI: 10.1021/acschemneuro.0c00244] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
![]()
Increasing human
life expectancy prompts the development of novel
remedies for cognitive decline: 44 million people worldwide are affected
by dementia, and this number is predicted to triple by 2050. Acetylcholinesterase
and N-methyl-d-aspartate receptors represent
the targets of currently available drugs for Alzheimer’s disease,
which are characterized by limited efficacy. Thus, the search for
therapeutic agents with alternative or combined mechanisms of action
is wide open. Since variations in 3′,5′-cyclic adenosine
monophosphate, 3′,5′-cyclic guanosine monophosphate,
and/or nitric oxide levels interfere with downstream pathways involved
in memory processes, evidence supporting the potential of phosphodiesterase
(PDE) inhibitors in contrasting neurodegeneration should be
critically considered. For the preparation of this Review, more than
140 scientific papers were retrieved by searching PubMed and Scopus
databases. A systematic approach was adopted when overviewing the
different PDE isoforms, taking into account details on brain localization,
downstream molecular mechanisms, and inhibitors currently under study,
according to available in vitro and in vivo data. In the context of drug repurposing, a section focusing on
PDE5 was introduced. Original computational studies were performed
to rationalize the emerging evidence that suggests the role of PDE5
inhibitors as multi-target agents against neurodegeneration.
Moreover, since such compounds must cross the blood–brain barrier
and reach inhibitory concentrations in the central nervous system
to exert their therapeutic activity, physicochemical parameters
were analyzed and discussed. Taken together, literature and computational
data suggest that some PDE5 inhibitors, such as tadalafil, represent
promising candidates.
Collapse
Affiliation(s)
- Giovanni Ribaudo
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Alberto Ongaro
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Giuseppe Zagotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Alessandra Gianoncelli
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| |
Collapse
|
41
|
Peng T, Qi B, He J, Ke H, Shi J. Advances in the Development of Phosphodiesterase-4 Inhibitors. J Med Chem 2020; 63:10594-10617. [PMID: 32255344 DOI: 10.1021/acs.jmedchem.9b02170] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cyclic nucleotide phosphodiesterase 4 (PDE4) specifically hydrolyzes cyclic adenosine monophosphate (cAMP) and plays vital roles in biological processes such as cancer development. To date, PDE4 inhibitors have been widely studied as therapeutics for the treatment of various diseases such as chronic obstructive pulmonary disease, and many of them have progressed to clinical trials or have been approved as drugs. Herein, we review the advances in the development of PDE4 inhibitors in the past decade and will focus on their pharmacophores, PDE4 subfamily selectivity, and therapeutic potential. Hopefully, this analysis will lead to a strategy for development of novel therapeutics targeting PDE4.
Collapse
Affiliation(s)
- Ting Peng
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Baowen Qi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jun He
- Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Sichuan 610041, China
| | - Hengming Ke
- Department of Biochemistry and Biophysics, and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu 610072, China
| |
Collapse
|
42
|
Morè L, Lauterborn JC, Papaleo F, Brambilla R. Enhancing cognition through pharmacological and environmental interventions: Examples from preclinical models of neurodevelopmental disorders. Neurosci Biobehav Rev 2020; 110:28-45. [PMID: 30981451 DOI: 10.1016/j.neubiorev.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/29/2022]
Abstract
In this review we discuss the role of environmental and pharmacological treatments to enhance cognition with special regards to neurodevelopmental related disorders and aging. How the environment influences brain structure and function, and the interactions between rearing conditions and gene expression, are fundamental questions that are still poorly understood. We propose a model that can explain some of the discrepancies in findings for effects of environmental enrichment on outcome measures. Evidence of a direct causal correlation of nootropics and treatments that enhanced cognition also will be presented, and possible molecular mechanisms that include neurotrophin signaling and downstream pathways underlying these processes are discussed. Finally we review recent findings achieved with a wide set of behavioral and cognitive tasks that have translational validity to humans, and should be useful for future work on devising appropriate therapies. As will be discussed, the collective findings suggest that a combinational therapeutic approach of environmental enrichment and nootropics could be particularly successful for improving learning and memory in both developmental disorders and normal aging.
Collapse
Affiliation(s)
- Lorenzo Morè
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, PR1 2XT, Preston, UK.
| | - Julie C Lauterborn
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92617, USA.
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163, Genova, Italy.
| | - Riccardo Brambilla
- Neuroscience and Mental Health Research Institute (NMHRI), Division of Neuroscience, School of Biosciences, Cardiff University, CF24 4HQ, Cardiff, UK.
| |
Collapse
|
43
|
Understanding PDE4's function in Alzheimer's disease; a target for novel therapeutic approaches. Biochem Soc Trans 2020; 47:1557-1565. [PMID: 31642904 PMCID: PMC6824677 DOI: 10.1042/bst20190763] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022]
Abstract
Phosphodiesterases (PDEs) have long been considered as targets for the treatment of Alzheimer's disease (AD) and a substantial body of evidence suggests that one sub-family from the super-family of PDEs, namely PDE4D, has particular significance in this context. This review discusses the role of PDE4 in the orchestration of cAMP response element binding signaling in AD and outlines the benefits of targeting PDE4D specifically. We examine the limited available literature that suggests PDE4 expression does not change in AD brains together with reports that show PDE4 inhibition as an effective treatment in this age-related neurodegenerative disease. Actually, aging induces changes in PDE4 expression/activity in an isoform and brain-region specific manner that proposes a similar complexity in AD brains. Therefore, a more detailed account of AD-related alterations in cellular/tissue location and the activation status of PDE4 is required before novel therapies can be developed to target cAMP signaling in this disease.
Collapse
|
44
|
Cavalloro V, Russo K, Vasile F, Pignataro L, Torretta A, Donini S, Semrau MS, Storici P, Rossi D, Rapetti F, Brullo C, Parisini E, Bruno O, Collina S. Insight into GEBR-32a: Chiral Resolution, Absolute Configuration and Enantiopreference in PDE4D Inhibition. Molecules 2020; 25:E935. [PMID: 32093112 PMCID: PMC7070305 DOI: 10.3390/molecules25040935] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/12/2020] [Accepted: 02/16/2020] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease is the most common type of dementia, affecting millions of people worldwide. One of its main consequences is memory loss, which is related to downstream effectors of cyclic adenosine monophosphate (cAMP). A well-established strategy to avoid cAMP degradation is the inhibition of phosphodiesterase (PDE). In recent years, GEBR-32a has been shown to possess selective inhibitory properties against PDE type 4 family members, resulting in an improvement in spatial memory processes without the typical side effects that are usually correlated with this mechanism of action. In this work, we performed the HPLC chiral resolution and absolute configuration assignment of GEBR-32a. We developed an efficient analytical and semipreparative chromatographic method exploiting an amylose-based stationary phase, we studied the chiroptical properties of both enantiomers and we assigned their absolute configuration by 1H-NMR (nuclear magnetic resonance). Lastly, we measured the IC50 values of both enantiomers against both the PDE4D catalytic domain and the long PDE4D3 isoform. Results strongly support the notion that GEBR-32a inhibits the PDE4D enzyme by interacting with both the catalytic pocket and the regulatory domains.
Collapse
Affiliation(s)
- Valeria Cavalloro
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
- Department of Earth and Environmental Sciences, University of Pavia, 27100 Pavia, Italy
| | - Katia Russo
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Francesca Vasile
- Università degli Studi di Milano, Dipartimento di Chimica, via C. Golgi 19, 20133 Milano, Italy
| | - Luca Pignataro
- Università degli Studi di Milano, Dipartimento di Chimica, via C. Golgi 19, 20133 Milano, Italy
| | - Archimede Torretta
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, Via Pascoli 70/3, 20133 Milano, Italy
| | - Stefano Donini
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, Via Pascoli 70/3, 20133 Milano, Italy
| | - Marta S Semrau
- Elettra-Sincrotrone Trieste S.C.p.A., SS 14-km 163.5 in AREA Science Park, 34149 Trieste, Italy
| | - Paola Storici
- Elettra-Sincrotrone Trieste S.C.p.A., SS 14-km 163.5 in AREA Science Park, 34149 Trieste, Italy
| | - Daniela Rossi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Federica Rapetti
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy
| | - Emilio Parisini
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, Via Pascoli 70/3, 20133 Milano, Italy
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Olga Bruno
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
45
|
Brullo C, Massa M, Rapetti F, Alfei S, Bertolotto MB, Montecucco F, Signorello MG, Bruno O. New Hybrid Pyrazole and Imidazopyrazole Antinflammatory Agents Able to Reduce ROS Production in Different Biological Targets. Molecules 2020; 25:899. [PMID: 32085423 PMCID: PMC7070443 DOI: 10.3390/molecules25040899] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 12/21/2022] Open
Abstract
Several anti-inflammatory agents based on pyrazole and imidazopyrazole scaffolds and a large library of substituted catechol PDE4D inhibitors were reported by us in the recent past. To obtain new molecules potentially able to act on different targets involved in inflammation onset we designed and synthesized a series of hybrid compounds by linking pyrazole and imidazo-pyrazole scaffolds to differently decorated catechol moieties through an acylhydrazone chain. Some compounds showed antioxidant activity, inhibiting reactive oxygen species (ROS) elevation in neutrophils, and a good inhibition of phosphodiesterases type 4D and, particularly, type 4B, the isoform most involved in inflammation. In addition, most compounds inhibited ROS production also in platelets, confirming their ability to exert an antiinflammatory response by two independent mechanism. Structure-activity relationship (SAR) analyses evidenced that both heterocyclic scaffolds (pyrazole and imidazopyrazole) and the substituted catechol moiety were determinant for the pharmacodynamic properties, even if hybrid molecules bearing to the pyrazole series were more active than the imidazopyrazole ones. In addition, the pivotal role of the catechol substituents has been analyzed. In conclusion the hybridization approach gave a new serie of multitarget antiinflammatory compounds, characterized by a strong antioxidant activity in different biological targets.
Collapse
Affiliation(s)
- Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, Viale Benedetto XV 3, I-16132 Genova, Italy; (M.M.); (F.R.); (O.B.)
| | - Matteo Massa
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, Viale Benedetto XV 3, I-16132 Genova, Italy; (M.M.); (F.R.); (O.B.)
| | - Federica Rapetti
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, Viale Benedetto XV 3, I-16132 Genova, Italy; (M.M.); (F.R.); (O.B.)
| | - Silvana Alfei
- Department of Pharmacy, Section of Chemistry and Pharmaceutical and Food Technologies, University of Genoa, Viale Cembrano 4, I-16148 Genova, Italy;
| | - Maria B. Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, and IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Largo R. Benzi 10, I-16132 Genoa, Italy; (M.B.B.); (F.M.)
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, and IRCCS Ospedale Policlinico San Martino Genova-Italian Cardiovascular Network, Largo R. Benzi 10, I-16132 Genoa, Italy; (M.B.B.); (F.M.)
| | - Maria Grazia Signorello
- Department of Pharmacy, Biochemistry Lab., University of Genoa, Viale Benedetto XV 3, I-16132 Genova, Italy;
| | - Olga Bruno
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, Viale Benedetto XV 3, I-16132 Genova, Italy; (M.M.); (F.R.); (O.B.)
| |
Collapse
|
46
|
Argyrousi EK, Heckman PRA, Prickaerts J. Role of cyclic nucleotides and their downstream signaling cascades in memory function: Being at the right time at the right spot. Neurosci Biobehav Rev 2020; 113:12-38. [PMID: 32044374 DOI: 10.1016/j.neubiorev.2020.02.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/23/2020] [Accepted: 02/03/2020] [Indexed: 01/23/2023]
Abstract
A plethora of studies indicate the important role of cAMP and cGMP cascades in neuronal plasticity and memory function. As a result, altered cyclic nucleotide signaling has been implicated in the pathophysiology of mnemonic dysfunction encountered in several diseases. In the present review we provide a wide overview of studies regarding the involvement of cyclic nucleotides, as well as their upstream and downstream molecules, in physiological and pathological mnemonic processes. Next, we discuss the regulation of the intracellular concentration of cyclic nucleotides via phosphodiesterases, the enzymes that degrade cAMP and/or cGMP, and via A-kinase-anchoring proteins that refine signal compartmentalization of cAMP signaling. We also provide an overview of the available data pointing to the existence of specific time windows in cyclic nucleotide signaling during neuroplasticity and memory formation and the significance to target these specific time phases for improving memory formation. Finally, we highlight the importance of emerging imaging tools like Förster resonance energy transfer imaging and optogenetics in detecting, measuring and manipulating the action of cyclic nucleotide signaling cascades.
Collapse
Affiliation(s)
- Elentina K Argyrousi
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands
| | - Pim R A Heckman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, 6200 MD, the Netherlands.
| |
Collapse
|
47
|
Blokland A, Heckman P, Vanmierlo T, Schreiber R, Paes D, Prickaerts J. Phosphodiesterase Type 4 Inhibition in CNS Diseases. Trends Pharmacol Sci 2019; 40:971-985. [DOI: 10.1016/j.tips.2019.10.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 12/17/2022]
|
48
|
Nabavi SM, Talarek S, Listos J, Nabavi SF, Devi KP, Roberto de Oliveira M, Tewari D, Argüelles S, Mehrzadi S, Hosseinzadeh A, D'onofrio G, Orhan IE, Sureda A, Xu S, Momtaz S, Farzaei MH. Phosphodiesterase inhibitors say NO to Alzheimer's disease. Food Chem Toxicol 2019; 134:110822. [PMID: 31536753 DOI: 10.1016/j.fct.2019.110822] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 09/13/2019] [Accepted: 09/14/2019] [Indexed: 12/18/2022]
Abstract
Phosphodiesterases (PDEs) consisted of 11 subtypes (PDE1 to PDE11) and over 40 isoforms that regulate levels of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP), the second messengers in cell functions. PDE inhibitors (PDEIs) have been attractive therapeutic targets due to their involvement in diverse medical conditions, e.g. cardiovascular diseases, autoimmune diseases, Alzheimer's disease (AD), etc. Among them; AD with a complex pathology is a progressive neurodegenerative disorder which affect mostly senile people in the world and only symptomatic treatment particularly using cholinesterase inhibitors in clinic is available at the moment for AD. Consequently, novel treatment strategies towards AD are still searched extensively. Since PDEs are broadly expressed in the brain, PDEIs are considered to modulate neurodegenerative conditions through regulating cAMP and cGMP in the brain. In this sense, several synthetic or natural molecules inhibiting various PDE subtypes such as rolipram and roflumilast (PDE4 inhibitors), vinpocetine (PDE1 inhibitor), cilostazol and milrinone (PDE3 inhibitors), sildenafil and tadalafil (PDE5 inhibitors), etc have been reported showing encouraging results for the treatment of AD. In this review, PDE superfamily will be scrutinized from the view point of structural features, isoforms, functions and pharmacology particularly attributed to PDEs as target for AD therapy.
Collapse
Affiliation(s)
- Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Sylwia Talarek
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St, 20-093, Lublin, Poland.
| | - Joanna Listos
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodźki 4a St, 20-093, Lublin, Poland.
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University, Karaikudi, 630003, Tamil Nadu, India.
| | - Marcos Roberto de Oliveira
- Departamento de Química (DQ), Instituto de Ciências Exatas e da Terra (ICET), Universidade Federal de Mato Grosso (UFMT), Cuiabá, Brazil.
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India.
| | - Sandro Argüelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Grazia D'onofrio
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS "Casa Sollievo della Sofferenza", Viale Cappuccini 1, 71013, San Giovanni Rotondo, FG, Italy.
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey.
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands, CIBEROBN (Physiopathology of Obesity and Nutrition), E-07122, Palma de Mallorca, Balearic Islands, Spain.
| | - Suowen Xu
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, 14623, USA.
| | - Saeedeh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
49
|
Pan T, Xie S, Zhou Y, Hu J, Luo H, Li X, Huang L. Dual functional cholinesterase and PDE4D inhibitors for the treatment of Alzheimer’s disease: Design, synthesis and evaluation of tacrine-pyrazolo[3,4-b]pyridine hybrids. Bioorg Med Chem Lett 2019; 29:2150-2152. [DOI: 10.1016/j.bmcl.2019.06.056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/21/2019] [Accepted: 06/28/2019] [Indexed: 10/26/2022]
|
50
|
Niego A, Benítez-Burraco A. Williams Syndrome, Human Self-Domestication, and Language Evolution. Front Psychol 2019; 10:521. [PMID: 30936846 PMCID: PMC6431629 DOI: 10.3389/fpsyg.2019.00521] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/22/2019] [Indexed: 01/06/2023] Open
Abstract
Language evolution resulted from changes in our biology, behavior, and culture. One source of these changes might be human self-domestication. Williams syndrome (WS) is a clinical condition with a clearly defined genetic basis which results in a distinctive behavioral and cognitive profile, including enhanced sociability. In this paper we show evidence that the WS phenotype can be satisfactorily construed as a hyper-domesticated human phenotype, plausibly resulting from the effect of the WS hemideletion on selected candidates for domestication and neural crest (NC) function. Specifically, we show that genes involved in animal domestication and NC development and function are significantly dysregulated in the blood of subjects with WS. We also discuss the consequences of this link between domestication and WS for our current understanding of language evolution.
Collapse
Affiliation(s)
- Amy Niego
- Ph.D. Program, Faculty of Humanities, University of Huelva, Huelva, Spain
| | - Antonio Benítez-Burraco
- Department of Spanish, Linguistics, and Theory of Literature, Faculty of Philology, University of Seville, Seville, Spain
| |
Collapse
|