1
|
Soon CW, Gaurav A, Gautam V, Al-Nema M. Structural insight into the lead identification of a dual inhibitor of PDE1B and PDE10A: Integrating pharmacophore-based virtual screening, molecular docking, and structure-activity-relationship approaches. Heliyon 2024; 10:e38305. [PMID: 39391487 PMCID: PMC11466560 DOI: 10.1016/j.heliyon.2024.e38305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 09/05/2024] [Accepted: 09/22/2024] [Indexed: 10/12/2024] Open
Abstract
Schizophrenia is a chronic neuropsychiatric disorder affecting more than 1% of the world's population. Current antipsychotic treatments show inadequacy in mitigating the negative and cognitive symptoms of schizophrenia. In addition, these medications cause undesirable extrapyramidal side effects. According to the studies, inhibition of phosphodiesterase (PDE) 1B and PDE10A simultaneously can alleviate positive, negative, and cognitive symptoms of schizophrenia. Thus, this study aims to identify new dual inhibitors of PDE1B and PDE10A using ligand-based pharmacophore modelling, virtual screening, and molecular docking studies. Accordingly, the generated pharmacophore models of PDE1B and PDE10A comprised hydrogen bond acceptor, aromatic ring, and hydrophobic features. These features were essential for retrieving the active hits from the Universal Natural Product Database in the virtual screening. Additional filters were subsequently employed to identify potential hits that could be developed into central nervous system-active compounds. Hits meeting all the screening criteria were subjected to docking studies with PDE1B and PDE10A. Among these hits, UNPD167314 exhibited significant binding affinities for the target receptors. It occupied the P-clamp and displayed hydrophobic, aromatic, and hydrogen bond interactions with the active site residues of both receptors, thus selected as a lead compound for the design of potent and selective dual inhibitors. The structural modifications of UNPD167314 resulted in the design of 35 novel inhibitors. Out of 35, four compounds exhibited high and comparable binding affinities for both PDE1B and PDE10A, making them promising candidates for further evaluation and optimisation.
Collapse
Affiliation(s)
- Ching Wen Soon
- Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Anand Gaurav
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248007, Uttarakhand, India
- Faculty of Health Sciences, Villa College, Male', 20373, Maldives
| | - Vertika Gautam
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India
| | - Mayasah Al-Nema
- Faculty of Pharmaceutical Sciences, UCSI University, Taman Connaught, Cheras, Kuala Lumpur, 56000, Malaysia
- Klarity, Westbourne, Bournemouth, BH4 8DT, United Kingdom
| |
Collapse
|
2
|
Ma R, Song N, Wang L, Gu X, Xiong F, Zhang S, Zhang J, Yang W, Zuo Z. Discovery of 2-(Methylcarbonylamino) thiazole as PDE4 inhibitors via virtual screening and biological evaluation. J Mol Graph Model 2023; 124:108567. [PMID: 37481883 DOI: 10.1016/j.jmgm.2023.108567] [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: 03/10/2023] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Phosphodiesterase-4, the primary enzyme responsible for cAMP degradation in the majority of immune and inflammatory cells, plays a critical role in the regulation of intracellular cAMP levels. Consequently, small molecular entities capable of inhibiting PDE4 have been employed in the treatment of inflammation-associated disorders, such as chronic obstructive pulmonary disease (COPD), psoriasis, atopic dermatitis (AD), inflammatory bowel diseases (IBD), rheumatic arthritis (RA). In the present investigation, a multi-faceted approach was employed to identify novel PDE4 inhibitors, utilizing the co-crystallization structure of PDE4B available in the Protein Data Bank (PDB) database, drug-like screening, false positive filtration, similarity and ADMET screen, as well as molecular docking via multiple software platforms, in conjunction with bioactivity assays. A thiazol-3-propanamides derivative, designated MR9, was discovered to inhibit PDE4B activity with IC50 values of 2.12 μM and suppress cellular inflammatory factor TNF-α release with an EC50 value of 3.587 μM. These findings suggest that the innovative active scaffold of MR9 offers a promising foundation for further structural refinement aimed at developing more potent PDE4 inhibitors.
Collapse
Affiliation(s)
- Rui Ma
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Na Song
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, PR China
| | - Lveli Wang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, PR China
| | - Xi Gu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, PR China
| | - Feng Xiong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, PR China
| | - Shuqun Zhang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, PR China
| | - Jie Zhang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, PR China
| | - Weimin Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, PR China.
| | - Zhili Zuo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| |
Collapse
|
3
|
Fernandes GFS, Lopes JR, Dos Santos JL, Scarim CB. Phthalimide as a versatile pharmacophore scaffold: Unlocking its diverse biological activities. Drug Dev Res 2023; 84:1346-1375. [PMID: 37492986 DOI: 10.1002/ddr.22094] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/27/2023]
Abstract
Phthalimide, a pharmacophore exhibiting diverse biological activities, holds a prominent position in medicinal chemistry. In recent decades, numerous derivatives of phthalimide have been synthesized and extensively studied for their therapeutic potential across a wide range of health conditions. This comprehensive review highlights the latest developments in medicinal chemistry, specifically focusing on phthalimide-based compounds that have emerged within the last decade. These compounds showcase promising biological activities, including anti-inflammatory, anti-Alzheimer, antiepileptic, antischizophrenia, antiplatelet, anticancer, antibacterial, antifungal, antimycobacterial, antiparasitic, anthelmintic, antiviral, and antidiabetic properties. The physicochemical profiles of the phthalimide derivatives were carefully analyzed using the online platform pkCSM, revealing the remarkable versatility of this scaffold. Therefore, this review emphasizes the potential of phthalimide as a valuable scaffold for the development of novel therapeutic agents, providing avenues for the exploration and design of new compounds.
Collapse
Affiliation(s)
| | - Juliana R Lopes
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Jean L Dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Cauê B Scarim
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
4
|
Abdallah MS, Mosalam EM, Hassan A, Ramadan AN, Omara‐Reda H, Zidan AA, Samman WA, El‐berri EI. Pentoxifylline as an adjunctive in treatment of negative symptoms in chronic schizophrenia: A double-blind, randomized, placebo-controlled trial. CNS Neurosci Ther 2023; 29:354-364. [PMID: 36341700 PMCID: PMC9804082 DOI: 10.1111/cns.14010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/03/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
Abstract
AIM The aim of this study was to explore the effectiveness and safety of pentoxifylline as an adjuvant to risperidone in mitigating the negative symptoms in patients with chronic schizophrenia. METHODS In this randomized, placebo-controlled study, eighty outpatients with chronic schizophrenia were given risperidone for 8 weeks along with either pentoxifylline or a placebo. The positive and negative syndrome scale (PANSS) was used to assess patients at the start of the trial, as well as at 2, 4, 6, and 8 weeks. Pre- and posttreatment serum levels of cAMP, TNF-α-, and IL-6 were measured. RESULTS The pentoxifylline group revealed a significant effect for time-treatment interaction on PANSS-negative subscale scores (p < 0.001), PANSS general psychopathology subscale scores (p < 0.001), and PANSS total scores (p < 0.001), but not on PANSS-positive subscale scores (p = 0.169). Additionally, when compared to the placebo group, the pentoxifylline group demonstrated a statistically significant increase in cAMP serum level and a statistically significant decrease in TNF-α and IL-6 serum levels. CONCLUSION Pentoxifylline adjunctive therapy with risperidone for 8 weeks was found to be promising in mitigating the negative symptoms in patients with chronic schizophrenia. TRIAL REGISTRATION NUMBER NCT04094207.
Collapse
Affiliation(s)
- Mahmoud S. Abdallah
- Department of Clinical Pharmacy, Faculty of PharmacyUniversity of Sadat CitySadat CityEgypt
| | - Esraa M. Mosalam
- Department of Biochemistry, Faculty of PharmacyMenoufia UniversityShebeen El‐KomEgypt
| | - Ahmed Hassan
- Department of Clinical Pharmacy, Faculty of PharmacyUniversity of Sadat CitySadat CityEgypt
| | - Ahmed N. Ramadan
- Department of Neuropsychiatry, Faculty of MedicineMenoufia UniversityShebeen El‐KomEgypt
| | - Hend Omara‐Reda
- Department of Neuropsychiatry, Faculty of MedicineMenoufia UniversityShebeen El‐KomEgypt
| | | | - Waad A. Samman
- Department of Pharmacology and Toxicology, College of PharmacyTaibah UniversityMedinaSaudi Arabia
| | - Eman I. El‐berri
- Clinical Pharmacy Department, Faculty of PharmacyTanta UniversityTantaEgypt
| |
Collapse
|
5
|
Non-Selective PDE4 Inhibition Induces a Rapid and Transient Decrease of Serum Potassium in Mice. BIOLOGY 2022; 11:biology11111582. [PMID: 36358283 PMCID: PMC9687940 DOI: 10.3390/biology11111582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Simple Summary Inhibitors of phosphodiesterase 4 (PDE4), a group of isoenzymes that hydrolyze and inactivate the second messenger cAMP, produce promising therapeutic benefits, including anti-inflammatory and memory-enhancing effects. Here, we report that, unexpectedly, PDE4 inhibitors also reduce serum potassium levels in mice. As both the total potassium content of the body, as well as the distribution of potassium between intra- and extracellular compartments, are critical for normal cellular functions, we further explored this observation. Several structurally distinct PDE4 inhibitors reduce serum potassium levels in mice, suggesting it is a class effect of these drugs. Serum potassium levels decrease within 15 min of drug injection, suggesting that PDE4 inhibition lowers serum potassium levels by promoting a transcellular shift of potassium from the blood into cells. This shift is a characteristically fast process, compared to a loss of total-body potassium via the kidneys or digestive tract (e.g., diarrhea). Indeed, stimulating cAMP synthesis with β-adrenoceptor agonists is known to rapidly shift potassium into cells, and PDE4 inhibitors appear to mimic this process by preventing PDE4-mediated cAMP degradation. Our findings reveal that the various acute physiologic effects of PDE4 inhibitors are paralleled and/or may be affected by reduced serum potassium levels. Abstract The analysis of blood samples from mice treated with the PDE4 inhibitor Roflumilast revealed an unexpected reduction in serum potassium levels, while sodium and chloride levels were unaffected. Treatment with several structurally distinct PAN-PDE4 inhibitors, including Roflumilast, Rolipram, RS25344, and YM976 dose-dependently reduced serum potassium levels, indicating the effect is a class-characteristic property. PDE4 inhibition also induces hypothermia and hypokinesia in mice. However, while general anesthesia abrogates these effects of PDE4 inhibitors, potassium levels decrease to similar extents in both awake as well as in fully anesthetized mice. This suggests that the hypokalemic effects of PDE4 inhibitors occur independently of hypothermia and hypokinesia. PDE4 inhibition reduces serum potassium within 15 min of treatment, consistent with a rapid transcellular shift of potassium. Catecholamines promote the uptake of potassium into the cell via increased cAMP signaling. PDE4 appears to modulate these adrenoceptor-mediated effects, as PDE4 inhibition has no additional effects on serum potassium in the presence of saturating doses of the β-adrenoceptor agonist Isoprenaline or the α2-blocker Yohimbine, and is partially blocked by pre-treatment with the β-blocker Propranolol. Together, these data suggest that PDE4 inhibitors reduce serum potassium levels by modulating the adrenergic regulation of cellular potassium uptake.
Collapse
|
6
|
Hazani R, Lavidor M, Weller A. Treatments for Social Interaction Impairment in Animal Models of Schizophrenia: A Critical Review and Meta-analysis. Schizophr Bull 2022; 48:1179-1193. [PMID: 35925025 PMCID: PMC9673263 DOI: 10.1093/schbul/sbac093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND While pharmacological treatments for positive symptoms of schizophrenia are widely used, their beneficial effect on negative symptoms, particularly social impairment, is insufficiently studied. Therefore, there is an increasing interest in preclinical research of potentially beneficial treatments, with mixed results. The current review aims to evaluate the efficacy of available treatments for social deficits in different animal models of schizophrenia. STUDY DESIGN A systematic literature search generated 145 outcomes for the measures "total time" and "number" of social interactions. Standardized mean differences (SMD) and 95% confidence interval (CI) were calculated, and heterogeneity was tested using Q statistics in a random-effect meta-analytic model. Given the vast heterogeneity in effect sizes, the animal model, treatment group, and sample size were all examined as potential moderators. STUDY RESULTS The results showed that in almost all models, treatment significantly improved social deficit (total time: SMD = 1.24; number: SMD = 1.1). The moderator analyses discovered significant subgroup differences across models and treatment subgroups. Perinatal and adult pharmacological models showed the most substantial influence of treatments on social deficits, reflecting relative pharmacological validity. Furthermore, atypical antipsychotic drugs had the highest SMD within each model subgroup. CONCLUSIONS Our findings indicate that the improvement in social interaction behaviors is dependent on the animal model and treatment family used. Implications for the preclinical and clinical fields are discussed.
Collapse
Affiliation(s)
- Reut Hazani
- To whom correspondence should be addressed; Department of Psychology, Bar-Ilan University, Ramat-Gan 5290002, Israel; tel: 972-3-531-8548, fax: 972-3-738-4173, e-mail:
| | - Michal Lavidor
- Psychology Department and Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Aron Weller
- Psychology Department and Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
7
|
Future Prospects of Positron Emission Tomography–Magnetic Resonance Imaging Hybrid Systems and Applications in Psychiatric Disorders. Pharmaceuticals (Basel) 2022; 15:ph15050583. [PMID: 35631409 PMCID: PMC9147426 DOI: 10.3390/ph15050583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/15/2022] Open
Abstract
A positron emission tomography (PET)–magnetic resonance imaging (MRI) hybrid system has been developed to improve the accuracy of molecular imaging with structural imaging. However, the mismatch in spatial resolution between the two systems hinders the use of the hybrid system. As the magnetic field of the MRI increased up to 7.0 tesla in the commercial system, the performance of the MRI system largely improved. Several technical attempts in terms of the detector and the software used with the PET were made to improve the performance. As a result, the high resolution of the PET–MRI fusion system enables quantitation of metabolism and molecular information in the small substructures of the brainstem, hippocampus, and thalamus. Many studies on psychiatric disorders, which are difficult to diagnose with medical imaging, have been accomplished using various radioligands, but only a few studies have been conducted using the PET–MRI fusion system. To increase the clinical usefulness of medical imaging in psychiatric disorders, a high-resolution PET–MRI fusion system can play a key role by providing important information on both molecular and structural aspects in the fine structures of the brain. The development of high-resolution PET–MR systems and their potential roles in clinical studies of psychiatric disorders were reviewed as prospective views in future diagnostics.
Collapse
|
8
|
Adenosine modulators and calcium channel blockers as add-on treatment for schizophrenia. NPJ SCHIZOPHRENIA 2021; 7:1. [PMID: 33479257 PMCID: PMC7820462 DOI: 10.1038/s41537-020-00135-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/24/2020] [Indexed: 11/30/2022]
Abstract
Relapses remain common among individuals with schizophrenia indicating a need for improved treatments. Creating a completely new drug molecule is expensive and time consuming, and therefore drug repurposing should be considered. Aim of this study was to investigate the risk of psychiatric rehospitalization associated with use of adenosine modulators (AMs) and calcium channel blockers (CCBs) in schizophrenia. Individuals diagnosed with schizophrenia (N = 61,889) in inpatient care between 1972–2014 in Finland were included. The follow-up lasted from 1996 to 2017. Main exposures were use of AMs (allopurinol and dipyridamole) and CCBs (dihydropyridines, diltiazem, and verapamil). Thiazide diuretics were used as a negative control. Within-individual models in stratified Cox regression were used and adjusted hazard ratios (HR) with 95% confidence intervals (CIs) are reported. Use of AMs was associated with a reduced risk of psychiatric rehospitalization on drug class level (HR 0.74, 95% CI 0.65–0.84, P < 0.0001), as well as on the level of individual drugs (allopurinol HR 0.82, 95% CI 0.70–0.97, P = 0.02; dipyridamole HR 0.65, 95% CI 0.55–0.77, P < 0.0001). Use of CCBs was associated with a reduced risk of psychiatric rehospitalization on drug class level (HR 0.81, 95% CI 0.77–0.86, P < 0.0001). From the different CCBs, only exposure to dihydropyridines was associated with a reduced risk (HR 0.79, 95% CI 0.74–0.84, P < 0.0001). No effect was observed for the negative control, thiazide diuretics (HR 0.96, 0.90–1.02, P = 0.20). The effects of dipyridamole and dihydropyridines were more pronounced among younger persons and combination of AMs, and CCBs was associated with a lower risk than either drug class as monotherapy. These results indicate a need for randomized controlled trials of these drugs.
Collapse
|
9
|
Amin HS, Parikh PK, Ghate MD. Medicinal chemistry strategies for the development of phosphodiesterase 10A (PDE10A) inhibitors - An update of recent progress. Eur J Med Chem 2021; 214:113155. [PMID: 33581555 DOI: 10.1016/j.ejmech.2021.113155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/27/2020] [Accepted: 01/03/2021] [Indexed: 11/26/2022]
Abstract
Phosphodiesterase 10A is a member of Phosphodiesterase (PDE)-superfamily of the enzyme which is responsible for hydrolysis of cAMP and cGMP to their inactive forms 5'-AMP and 5'-GMP, respectively. PDE10A is highly expressed in the brain, particularly in the putamen and caudate nucleus. PDE10A plays an important role in the regulation of localization, duration, and amplitude of the cyclic nucleotide signalling within the subcellular domain of these regions, and thereby modulation of PDE10A enzyme can give rise to a new therapeutic approach in the treatment of schizophrenia and other neurodegenerative disorders. Limitation of the conventional therapy of schizophrenia forced the pharmaceutical industry to move their efforts to develop a novel treatment approach with reduced side effects. In the past decade, considerable developments have been made in pursuit of PDE10A centric antipsychotic agents by several pharmaceutical industries due to the distribution of PDE10A in the brain and the ability of PDE10A inhibitors to mimic the effect of D2 antagonists and D1 agonists. However, no selective PDE10A inhibitor is currently available in the market for the treatment of schizophrenia. The present compilation concisely describes the role of PDE10A inhibitors in the therapy of neurodegenerative disorders mainly in psychosis, the structure of PDE10A enzyme, key interaction of different PDE10A inhibitors with human PDE10A enzyme and recent medicinal chemistry developments in designing of safe and effective PDE10A inhibitors for the treatment of schizophrenia. The present compilation also provides useful information and future direction to bring further improvements in the discovery of PDE10A inhibitors.
Collapse
Affiliation(s)
- Harsh S Amin
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India
| | - Palak K Parikh
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India; Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India.
| | - Manjunath D Ghate
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India
| |
Collapse
|
10
|
Piazza GA, Ward A, Chen X, Maxuitenko Y, Coley A, Aboelella NS, Buchsbaum DJ, Boyd MR, Keeton AB, Zhou G. PDE5 and PDE10 inhibition activates cGMP/PKG signaling to block Wnt/β-catenin transcription, cancer cell growth, and tumor immunity. Drug Discov Today 2020; 25:1521-1527. [PMID: 32562844 DOI: 10.1016/j.drudis.2020.06.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/10/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023]
Abstract
Although numerous reports conclude that nonsteroidal anti-inflammatory drugs (NSAIDs) have anticancer activity, this common drug class is not recommended for long-term use because of potentially fatal toxicities from cyclooxygenase (COX) inhibition. Studies suggest the mechanism responsible for the anticancer activity of the NSAID sulindac is unrelated to COX inhibition but instead involves an off-target, phosphodiesterase (PDE). Thus, it might be feasible develop safer and more efficacious drugs for cancer indications by targeting PDE5 and PDE10, which are overexpressed in various tumors and essential for cancer cell growth. In this review, we describe the rationale for using the sulindac scaffold to design-out COX inhibitory activity, while improving potency and selectivity to inhibit PDE5 and PDE10 that activate cGMP/PKG signaling to suppress Wnt/β-catenin transcription, cancer cell growth, and tumor immunity.
Collapse
Affiliation(s)
- Gary A Piazza
- Drug Discovery Research Center, Department of Pharmacology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA.
| | - Antonio Ward
- Drug Discovery Research Center, Department of Pharmacology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Xi Chen
- Drug Discovery Research Center, Department of Pharmacology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Yulia Maxuitenko
- Drug Discovery Research Center, Department of Pharmacology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Alex Coley
- Drug Discovery Research Center, Department of Pharmacology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | | | - Donald J Buchsbaum
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Adam B Keeton
- Drug Discovery Research Center, Department of Pharmacology, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| |
Collapse
|
11
|
Bieber M, Schuhmann MK, Volz J, Kumar GJ, Vaidya JR, Nieswandt B, Pham M, Stoll G, Kleinschnitz C, Kraft P. Description of a Novel Phosphodiesterase (PDE)-3 Inhibitor Protecting Mice From Ischemic Stroke Independent From Platelet Function. Stroke 2019; 50:478-486. [PMID: 30566040 PMCID: PMC6358218 DOI: 10.1161/strokeaha.118.023664] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Supplemental Digital Content is available in the text. Background and Purpose— Acetylsalicylic acid and clopidogrel are the 2 main antithrombotic drugs for secondary prevention in patients with ischemic stroke (IS) without indication for anticoagulation. Because of their limited efficacy and potential side effects, novel antiplatelet agents are urgently needed. Cilostazol, a specific phosphodiesterase (PDE)-3 inhibitor, protected from IS in clinical studies comprising mainly Asian populations. Nevertheless, the detailed mechanistic role of PDE-3 inhibitors in IS pathophysiology is hardly understood. In this project, we analyzed the efficacy and pathophysiologic mechanisms of a novel and only recently described PDE-3 inhibitor (substance V) in a mouse model of focal cerebral ischemia. Methods— Focal cerebral ischemia was induced by transient middle cerebral artery occlusion in 6- to 8-week-old male C57Bl/6 wild-type mice receiving substance V or vehicle 1 hour after ischemia induction. Infarct volumes and functional outcomes were assessed between day 1 and day 7, and findings were validated by magnetic resonance imaging. Blood-brain barrier damage, as well as the extent of local inflammatory response and cell death, was determined. Results— Inhibition of PDE-3 by pharmacological blockade with substance V significantly reduced infarct volumes and improved neurological outcome on day 1 and 7 after experimental cerebral ischemia. Reduced blood-brain barrier damage, attenuated brain tissue inflammation, and decreased local cell death could be identified as potential mechanisms. PDE-3 inhibitor treatment did neither increase the number of intracerebral hemorrhages nor affect platelet function. Conclusions— The novel PDE-3 inhibitor substance V protected mice from IS independent from platelet function. Pharmaceutical inactivation of PDE-3 might become a promising therapeutic approach to combat IS via inhibition of thromboinflammatory mechanisms and stabilization of the blood-brain barrier.
Collapse
Affiliation(s)
- Michael Bieber
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany
| | - Michael K Schuhmann
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany
| | - Julia Volz
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Germany (J.V., B.N.)
| | - Gangasani Jagadeesh Kumar
- Fluro Agro Chemicals (Organic Chemistry II) Division (G.J.K., J.R.V.) and AcSIR (G.J.K., J.R.V.), CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
| | - Jayathirtha Rao Vaidya
- Fluro Agro Chemicals (Organic Chemistry II) Division (G.J.K., J.R.V.) and AcSIR (G.J.K., J.R.V.), CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, India
| | - Bernhard Nieswandt
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Germany (J.V., B.N.)
| | - Mirko Pham
- Department of Neuroradiology (M.P.), University Hospital Würzburg, Germany
| | - Guido Stoll
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany
| | - Christoph Kleinschnitz
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany.,Department of Neurology, University Hospital Essen, Germany (C.K.)
| | - Peter Kraft
- From the Department of Neurology (M.B., M.K.S., G.S., C.K., P.K.), University Hospital Würzburg, Germany.,Department of Neurology, Klinikum Main-Spessart, Lohr, Germany (P.K.)
| |
Collapse
|
12
|
Czopek A, Bucki A, Kołaczkowski M, Zagórska A, Drop M, Pawłowski M, Siwek A, Głuch-Lutwin M, Pękala E, Chrzanowska A, Struga M, Partyka A, Wesołowska A. Novel multitarget 5-arylidenehydantoins with arylpiperazinealkyl fragment: Pharmacological evaluation and investigation of cytotoxicity and metabolic stability. Bioorg Med Chem 2019; 27:4163-4173. [DOI: 10.1016/j.bmc.2019.07.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/17/2019] [Accepted: 07/28/2019] [Indexed: 12/21/2022]
|
13
|
The role of striatum and prefrontal cortex in the prevention of amphetamine-induced schizophrenia-like effects mediated by nitric oxide compounds. Prog Neuropsychopharmacol Biol Psychiatry 2018; 86:353-362. [PMID: 29555252 DOI: 10.1016/j.pnpbp.2018.03.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/14/2018] [Accepted: 03/14/2018] [Indexed: 01/07/2023]
Abstract
Pharmacological manipulation of nitric oxide (NO) has been suggested as a promising treatment for schizophrenia symptoms. A single infusion of sodium nitroprusside, a NO donor with short half-life, was found to improve schizophrenia symptoms. However, an increasing number of preclinical studies have demonstrated the potential beneficial effects of both NO donors and inhibitors. We investigated the potential synergistic effect of sub-effective doses of the NO donor sodium nitroprusside or the NO inhibitor 7-Nitroindazole (7NI) combined with clozapine, a standard atypical antipsychotic, on counteracting amphetamine or MK-801-induced psychosis-like behaviors. The impact of sodium nitroprusside and 7NI on cAMP regulation in the prefrontal cortex and striatum was also evaluated. Confirming previous results, we found that both NO donors and inhibitors prevented amphetamine-induced effects (prepulse inhibition [PPI] disruption and hyperlocomotion). In addition, we observed a synergistic effect of sodium nitroprusside and clozapine on antagonizing the disruptive effects of amphetamine, but not MK-801, in the PPI test. The sub-effective dose of 7NI tested did not prevent amphetamine or MK-induced PPI effects when combined with clozapine. Interestingly, cAMP levels were significantly decreased in the prefrontal cortex after treatment with sodium nitroprusside. In the striatum, both sodium nitroprusside and 7NI blocked the amphetamine-induced increase of cAMP. Our data corroborate previous findings on the dopaminergic mechanisms involved in the action of sodium nitroprusside. It is likely that the differential effects of sodium nitroprusside are related to its ability to modify cAMP levels in the prefrontal cortex.
Collapse
|
14
|
Heckman PRA, Blokland A, Bollen EPP, Prickaerts J. Phosphodiesterase inhibition and modulation of corticostriatal and hippocampal circuits: Clinical overview and translational considerations. Neurosci Biobehav Rev 2018; 87:233-254. [PMID: 29454746 DOI: 10.1016/j.neubiorev.2018.02.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/07/2018] [Accepted: 02/09/2018] [Indexed: 12/20/2022]
Abstract
The corticostriatal and hippocampal circuits contribute to the neurobiological underpinnings of several neuropsychiatric disorders, including Alzheimer's disease, Parkinson's disease and schizophrenia. Based on biological function, these circuits can be clustered into motor circuits, associative/cognitive circuits and limbic circuits. Together, dysfunctions in these circuits produce the wide range of symptoms observed in related neuropsychiatric disorders. Intracellular signaling in these circuits is largely mediated through the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway with an additional role for the cyclic guanosine monophosphate (cGMP)/ protein kinase G (PKG) pathway, both of which can be regulated by phosphodiesterase inhibitors (PDE inhibitors). Through their effects on cAMP response element-binding protein (CREB) and Dopamine- and cAMP-Regulated PhosphoProtein MR 32 kDa (DARPP-32), cyclic nucleotide pathways are involved in synaptic transmission, neuron excitability, neuroplasticity and neuroprotection. In this clinical review, we provide an overview of the current clinical status, discuss the general mechanism of action of PDE inhibitors in relation to the corticostriatal and hippocampal circuits and consider several translational challenges.
Collapse
Affiliation(s)
- P R A Heckman
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands; Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, The Netherlands.
| | - A Blokland
- Department of Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, The Netherlands
| | - E P P Bollen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - J Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
15
|
Puigdellívol M, Saavedra A, Pérez-Navarro E. Cognitive dysfunction in Huntington's disease: mechanisms and therapeutic strategies beyond BDNF. Brain Pathol 2018; 26:752-771. [PMID: 27529673 DOI: 10.1111/bpa.12432] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/08/2016] [Indexed: 12/15/2022] Open
Abstract
One of the main focuses in Huntington's disease (HD) research, as well as in most neurodegenerative diseases, is the development of new therapeutic strategies, as currently there is no treatment to delay or prevent the progression of the disease. Neuronal dysfunction and neuronal death in HD are caused by a combination of interrelated pathogenic processes that lead to motor, cognitive and psychiatric symptoms. Understanding how mutant huntingtin impacts on a plethora of cellular functions could help to identify new molecular targets. Although HD has been classically classified as a neurodegenerative disease affecting voluntary movement, lately cognitive dysfunction is receiving increased attention as it is very invalidating for patients. Thus, an ambitious goal in HD research is to find altered molecular mechanisms that contribute to cognitive decline. In this review, we have focused on those findings related to corticostriatal and hippocampal cognitive dysfunction in HD, as well as on the underlying molecular mechanisms, which constitute potential therapeutic targets. These include alterations in synaptic plasticity, transcriptional machinery and neurotrophic and neurotransmitter signaling.
Collapse
Affiliation(s)
- Mar Puigdellívol
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red (CIBER) sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Ana Saavedra
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red (CIBER) sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Institut de Neurociències, Universitat de Barcelona, Catalonia, Spain
| | - Esther Pérez-Navarro
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Catalonia, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.,Centro de Investigación Biomédica en Red (CIBER) sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.,Institut de Neurociències, Universitat de Barcelona, Catalonia, Spain
| |
Collapse
|
16
|
Zhu B, Lindsey A, Li N, Lee K, Ramirez-Alcantara V, Canzoneri JC, Fajardo A, Madeira da Silva L, Thomas M, Piazza JT, Yet L, Eberhardt BT, Gurpinar E, Otali D, Grizzle W, Valiyaveettil J, Chen X, Keeton AB, Piazza GA. Phosphodiesterase 10A is overexpressed in lung tumor cells and inhibitors selectively suppress growth by blocking β-catenin and MAPK signaling. Oncotarget 2017; 8:69264-69280. [PMID: 29050202 PMCID: PMC5642477 DOI: 10.18632/oncotarget.20566] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/04/2017] [Indexed: 12/14/2022] Open
Abstract
Phosphodiesterase 10A (PDE10) is a cyclic nucleotide (e.g. cGMP) degrading enzyme highly expressed in the brain striatum where it plays an important role in dopaminergic neurotransmission, but has limited expression and no known physiological function outside the central nervous system. Here we report that PDE10 mRNA and protein levels are strongly elevated in human non-small cell lung cancer cells and lung tumors compared with normal human airway epithelial cells and lung tissue, respectively. Genetic silencing of PDE10 or inhibition by small molecules such as PQ10 was found to selectively inhibit the growth and colony formation of lung tumor cells. PQ10 treatment of lung tumor cells rapidly increased intracellular cGMP levels and activated cGMP-dependent protein kinase (PKG) at concentrations that inhibit lung tumor cell growth. PQ10 also increased the phosphorylation of β-catenin and reduced its levels, which paralleled the suppression of cyclin D1 and survivin but preceded the activation of PARP and caspase cleavage. PQ10 also suppressed RAS-activated RAF/MAPK signaling within the same concentration range and treatment period as required for cGMP elevation and PKG activation. These results show that PDE10 is overexpressed during lung cancer development and essential for lung tumor cell growth in which inhibitors can selectively induce apoptosis by increasing intracellular cGMP levels and activating PKG to suppress oncogenic β-catenin and MAPK signaling.
Collapse
Affiliation(s)
- Bing Zhu
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Ashley Lindsey
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Nan Li
- Department of Biochemistry and Molecular Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kevin Lee
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Veronica Ramirez-Alcantara
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Joshua C Canzoneri
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Alexandra Fajardo
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Luciana Madeira da Silva
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Meagan Thomas
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - John T Piazza
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Larry Yet
- Department of Chemistry, University of South Alabama, Mobile, Alabama, USA
| | - Brian T Eberhardt
- Department of Chemistry, University of South Alabama, Mobile, Alabama, USA
| | - Evrim Gurpinar
- Department of Pharmacology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dennis Otali
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - William Grizzle
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jacob Valiyaveettil
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Xi Chen
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Adam B Keeton
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Gary A Piazza
- Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
17
|
Mori W, Takei M, Furutsuka K, Fujinaga M, Kumata K, Muto M, Ohkubo T, Hashimoto H, Tamagnan G, Higuchi M, Kawamura K, Zhang MR. Comparison between [ 18F]fluorination and [ 18F]fluoroethylation reactions for the synthesis of the PDE10A PET radiotracer [ 18F]MNI-659. Nucl Med Biol 2017; 55:12-18. [PMID: 28972915 DOI: 10.1016/j.nucmedbio.2017.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/04/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022]
Abstract
INTRODUCTION 2-(2-(3-(4-(2-[18F]Fluoroethoxy)phenyl)-7-methyl-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)-4-isopropoxyisoindoline-1,3-dione ([18F]MNI-659, [18F]1) is a useful PET radiotracer for imaging phosphodiesterase 10A (PDE10A) in human brain. [18F]1 has been previously prepared by direct [18F]fluorination of a tosylate precursor 2 with [18F]F-. The aim of this study was to determine the conditions for the [18F]fluorination reaction to obtain [18F]1 of high quality and with sufficient radioactivity for clinical use in our institute. Moreover, we synthesized [18F]1 by [18F]fluoroethylation of a phenol precursor 3 with [18F]fluoroethyl bromide ([18F]FEtBr), and the outcomes of [18F]fluorination and [18F]fluoroethylation were compared. METHODS We performed the automated synthesis of [18F]1 by [18F]fluorination and [18F]fluoroethylation using a multi-purpose synthesizer. We determined the amounts of tosylate precursor 2 and potassium carbonate as well as the reaction temperature for direct [18F]fluorination. RESULTS The efficiency of the [18F]fluorination reaction was strongly affected by the amount of 2 and potassium carbonate. Under the determined reaction conditions, [18F]1 with 0.82±0.2GBq was obtained in 13.6%±3.3% radiochemical yield (n=8, decay-corrected to EOB and based on [18F]F-) at EOS, starting from 11.5±0.4GBq of cyclotron-produced [18F]F-. On the other hand, the [18F]fluoroethylation of 3 with [18F]FEtBr produced [18F]1 with 1.0±0.2GBq and in 22.5±2.5 % radiochemical yields (n=7, decay-corrected to EOB and based on [18F]F-) at EOS, starting from 7.4GBq of cyclotron-produced [18F]F-. Clearly, [18F]fluoroethylation resulted in a higher radiochemical yield of [18F]1 than [18F]fluorination. CONCLUSION [18F]1 of high quality and with sufficient radioactivity was successfully radiosynthesized by two methods. [18F]1 synthesized by direct [18F]fluorination has been approved and will be provided for clinical use in our institute.
Collapse
Affiliation(s)
- Wakana Mori
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Makoto Takei
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Kenji Furutsuka
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; SHI Accelerator Service Ltd., Tokyo 141-0032, Japan
| | - Masayuki Fujinaga
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Katsushi Kumata
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Masatoshi Muto
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; Tokyo Nuclear Services Ltd., Tokyo 110-0016, Japan
| | - Takayuki Ohkubo
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; SHI Accelerator Service Ltd., Tokyo 141-0032, Japan
| | - Hiroki Hashimoto
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | | | - Makoto Higuchi
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Kazunori Kawamura
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan.
| |
Collapse
|
18
|
Rezaei F, Mesgarpour B, Jeddian A, Zeionoddini A, Mohammadinejad P, Salardini E, Shahriari M, Zeinoddini A, Akhondzadeh S. Cilostazol adjunctive therapy in treatment of negative symptoms in chronic schizophrenia: Randomized, double-blind, placebo-controlled study. Hum Psychopharmacol 2017; 32. [PMID: 28421639 DOI: 10.1002/hup.2583] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 03/03/2017] [Accepted: 03/05/2017] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To evaluate the efficacy and safety of cilostazol, a selective inhibitor of phosphodiesterase III, as an adjunctive to risperidone in alleviating the negative symptoms of schizophrenia. METHODS Eighty-four in-patients with diagnosis of chronic schizophrenia participated in a randomized, placebo-controlled trial and underwent 8 weeks of treatment with either cilostazol (50 mg twice a day) or placebo as an adjuvant to risperidone. Participants were assessed using the positive and negative syndrome scale (PANSS) at baseline and at weeks 2, 4, 6, and 8. The primary outcome measure of the trial was to evaluate the efficacy of cilostazol compared to placebo in improving the PANSS negative subscale score. RESULT General linear model repeated measures demonstrated significant effect for time × treatment interaction on negative subscale scores (p < .001) and PANSS total (p = .006) but did not demonstrate significant effect on the PANSS positive (p = .37) and general (p = .06) subscales. Frequency of adverse events was not significantly different between the 2 treatment groups. No serious adverse event was observed. CONCLUSION An 8-week course of treatment with cilostazol as an adjunct to risperidone showed a favorable safety and efficacy profile in patients with schizophrenia.
Collapse
Affiliation(s)
- Farzin Rezaei
- Qods Hospital, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Bita Mesgarpour
- National Institute for Medical Research Development, Tehran, Iran
| | - Alireza Jeddian
- Digestive Disease Research Institute, Tehran University of medical science, Tehran, Iran
| | - Atefeh Zeionoddini
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Payam Mohammadinejad
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Elaheh Salardini
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Shahriari
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arefeh Zeinoddini
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Knott EP, Assi M, Rao SNR, Ghosh M, Pearse DD. Phosphodiesterase Inhibitors as a Therapeutic Approach to Neuroprotection and Repair. Int J Mol Sci 2017; 18:E696. [PMID: 28338622 PMCID: PMC5412282 DOI: 10.3390/ijms18040696] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 03/10/2017] [Accepted: 03/15/2017] [Indexed: 12/21/2022] Open
Abstract
A wide diversity of perturbations of the central nervous system (CNS) result in structural damage to the neuroarchitecture and cellular defects, which in turn are accompanied by neurological dysfunction and abortive endogenous neurorepair. Altering intracellular signaling pathways involved in inflammation and immune regulation, neural cell death, axon plasticity and remyelination has shown therapeutic benefit in experimental models of neurological disease and trauma. The second messengers, cyclic adenosine monophosphate (cyclic AMP) and cyclic guanosine monophosphate (cyclic GMP), are two such intracellular signaling targets, the elevation of which has produced beneficial cellular effects within a range of CNS pathologies. The only known negative regulators of cyclic nucleotides are a family of enzymes called phosphodiesterases (PDEs) that hydrolyze cyclic nucleotides into adenosine monophosphate (AMP) or guanylate monophosphate (GMP). Herein, we discuss the structure and physiological function as well as the roles PDEs play in pathological processes of the diseased or injured CNS. Further we review the approaches that have been employed therapeutically in experimental paradigms to block PDE expression or activity and in turn elevate cyclic nucleotide levels to mediate neuroprotection or neurorepair as well as discuss both the translational pathway and current limitations in moving new PDE-targeted therapies to the clinic.
Collapse
Affiliation(s)
- Eric P Knott
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Mazen Assi
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Sudheendra N R Rao
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Department of Neurological Surgery, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Department of Neurological Surgery, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Neuroscience Program, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, The Miller School of Medicine at the University of Miami, Miami, FL 33136, USA.
- Bruce Wayne Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA.
| |
Collapse
|
20
|
Ooms M, Attili B, Celen S, Koole M, Verbruggen A, Van Laere K, Bormans G. [18F]JNJ42259152 binding to phosphodiesterase 10A, a key regulator of medium spiny neuron excitability, is altered in the presence of cyclic AMP. J Neurochem 2016; 139:897-906. [DOI: 10.1111/jnc.13855] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/29/2016] [Accepted: 09/19/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Maarten Ooms
- Laboratory for Radiopharmacy; Department of Pharmaceutical and Pharmacological Sciences KU Leuven; Leuven Belgium
| | - Bala Attili
- Laboratory for Radiopharmacy; Department of Pharmaceutical and Pharmacological Sciences KU Leuven; Leuven Belgium
| | - Sofie Celen
- Laboratory for Radiopharmacy; Department of Pharmaceutical and Pharmacological Sciences KU Leuven; Leuven Belgium
| | - Michel Koole
- Division of Nuclear Medicine; KU Leuven and University Hospital Leuven; Leuven Belgium
| | - Alfons Verbruggen
- Laboratory for Radiopharmacy; Department of Pharmaceutical and Pharmacological Sciences KU Leuven; Leuven Belgium
| | - Koen Van Laere
- Division of Nuclear Medicine; KU Leuven and University Hospital Leuven; Leuven Belgium
| | - Guy Bormans
- Laboratory for Radiopharmacy; Department of Pharmaceutical and Pharmacological Sciences KU Leuven; Leuven Belgium
| |
Collapse
|
21
|
Goupil S, Maréchal L, El Hajj H, Tremblay MÈ, Richard FJ, Leclerc P. Identification and Localization of the Cyclic Nucleotide Phosphodiesterase 10A in Bovine Testis and Mature Spermatozoa. PLoS One 2016; 11:e0161035. [PMID: 27548062 PMCID: PMC4993467 DOI: 10.1371/journal.pone.0161035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/28/2016] [Indexed: 11/19/2022] Open
Abstract
In mammals, adenosine 3’, 5’-cyclic monophosphate (cAMP) is known to play highly important roles in sperm motility and acrosomal exocytosis. It is known to act through protein phosphorylation via PRKA and through the activation of guanine nucleotide exchange factors like EPAC. Sperm intracellular cAMP levels depend on the activity of adenylyl cyclases, mostly SACY, though transmembrane-containing adenylyl cyclases are also present, and on the activity of cyclic nucleotide phosphodiesterases (PDE) whose role is to degrade cAMP into 5’-AMP. The PDE superfamily is subdivided into 11 families (PDE1 to 11), which act on either cAMP or cGMP, or on both cAMP and cGMP although with different enzymatic properties. PDE10, which is more effective on cAMP than cGMP, has been known for almost 15 years and is mostly studied in the brain where it is associated with neurological disorders. Although a high level of PDE10A gene expression is observed in the testis, information on the identity of the isoforms or on the cell type that express the PDE10 protein is lacking. The objective of this study was to identify the PDE10A isoforms expressed in the testis and germ cells, and to determine the presence and localization of PDE10A in mature spermatozoa. As a sub-objective, since PDE10A transcript variants were reported strictly through analyses of bovine genomic sequence, we also wanted to determine the nucleotide and amino acid sequences by experimental evidence. Using RT-PCR, 5’- and 3’-RACE approaches we clearly show that PDE10A transcript variants X3 and X5 are expressed in bovine testis as well as in primary spermatocytes and spermatids. We also reveal using a combination of immunological techniques and proteomics analytical tools that the PDE10A isoform X4 is present in the area of the developing acrosome of spermatids and of the acrosome of mature spermatozoa.
Collapse
Affiliation(s)
- Serge Goupil
- Département d’obstétrique, gynécologie et reproduction, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
| | - Loïze Maréchal
- Département d’obstétrique, gynécologie et reproduction, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
| | - Hassan El Hajj
- Département de médecine moléculaire, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Département de médecine moléculaire, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
| | - François J. Richard
- Département des sciences animales, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Université Laval, Québec, Canada
| | - Pierre Leclerc
- Département d’obstétrique, gynécologie et reproduction, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
- Centre de recherche en reproduction, développement et santé intergénérationnelle (CRDSI), Université Laval, Québec, Canada
- Centre de recherche du CHU de Québec-Université Laval, G1V 4G2, Québec, QC, Canada
- * E-mail:
| |
Collapse
|
22
|
Bergman O, Ben-Shachar D. Mitochondrial Oxidative Phosphorylation System (OXPHOS) Deficits in Schizophrenia: Possible Interactions with Cellular Processes. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2016; 61:457-69. [PMID: 27412728 PMCID: PMC4959648 DOI: 10.1177/0706743716648290] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondria are key players in the generation and regulation of cellular bioenergetics, producing the majority of adenosine triphosphate molecules by the oxidative phosphorylation system (OXPHOS). Linked to numerous signaling pathways and cellular functions, mitochondria, and OXPHOS in particular, are involved in neuronal development, connectivity, plasticity, and differentiation. Impairments in a variety of mitochondrial functions have been described in different general and psychiatric disorders, including schizophrenia (SCZ), a severe, chronic, debilitating illness that heavily affects the lives of patients and their families. This article reviews findings emphasizing the role of OXPHOS in the pathophysiology of SCZ. Evidence accumulated during the past few decades from imaging, transcriptomic, proteomic, and metabolomic studies points at OXPHOS deficit involvement in SCZ. Abnormalities have been reported in high-energy phosphates generated by the OXPHOS, in the activity of its complexes and gene expression, primarily of complex I (CoI). In addition, cellular signaling such as cAMP/protein kinase A (PKA) and Ca(+2), neuronal development, connectivity, and plasticity have been linked to OXPHOS function and are reported to be impaired in SCZ. Finally, CoI has been shown as a site of interaction for both dopamine (DA) and antipsychotic drugs, further substantiating its role in the pathology of SCZ. Understanding the role of mitochondria and the OXPHOS in particular may encourage new insights into the pathophysiology and etiology of this debilitating disorder.
Collapse
Affiliation(s)
- Oded Bergman
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Medical Center, Technion-IIT, Haifa, Israel B. Rappaport Faculty of Medicine, Technion-IIT, Haifa, Israel
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Psychiatry, Rambam Medical Center, Technion-IIT, Haifa, Israel B. Rappaport Faculty of Medicine, Technion-IIT, Haifa, Israel
| |
Collapse
|
23
|
The effects of PDE10 inhibition on attentional set-shifting do not depend on the activation of dopamine D1 receptors. Behav Pharmacol 2016; 27:331-8. [DOI: 10.1097/fbp.0000000000000201] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
24
|
Abstract
Although there has been more than 50 years of development, there remains a great need for better antipsychotic medications. This article looks at the recent advances in treatment of schizophrenia. New hypotheses have been suggested that may replace or complement the dopamine hypotheses. The article explores the different novel drugs that impact some of the key neurotransmitter systems currently. Phosphodiesterase 10A inhibitors and α-7 neuronal nicotinic acetylcholine receptor modulators constitute the majority. The marketing of these medications eventually may result in change about how schizophrenia is treated.
Collapse
|
25
|
Heckman PRA, van Duinen MA, Bollen EPP, Nishi A, Wennogle LP, Blokland A, Prickaerts J. Phosphodiesterase Inhibition and Regulation of Dopaminergic Frontal and Striatal Functioning: Clinical Implications. Int J Neuropsychopharmacol 2016; 19:pyw030. [PMID: 27037577 PMCID: PMC5091819 DOI: 10.1093/ijnp/pyw030] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 03/30/2016] [Accepted: 03/30/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The fronto-striatal circuits are the common neurobiological basis for neuropsychiatric disorders, including schizophrenia, Parkinson's disease, Huntington's disease, attention deficit hyperactivity disorder, obsessive-compulsive disorder, and Tourette's syndrome. Fronto-striatal circuits consist of motor circuits, associative circuits, and limbic circuits. All circuits share 2 common features. First, all fronto-striatal circuits consist of hyper direct, direct, and indirect pathways. Second, all fronto-striatal circuits are modulated by dopamine. Intracellularly, the effect of dopamine is largely mediated through the cyclic adenosine monophosphate/protein kinase A signaling cascade with an additional role for the cyclic guanosine monophosphate/protein kinase G pathway, both of which can be regulated by phosphodiesterases. Phosphodiesterases are thus a potential target for pharmacological intervention in neuropsychiatric disorders related to dopaminergic regulation of fronto-striatal circuits. METHODS Clinical studies of the effects of different phosphodiesterase inhibitors on cognition, affect, and motor function in relation to the fronto-striatal circuits are reviewed. RESULTS Several selective phosphodiesterase inhibitors have positive effects on cognition, affect, and motor function in relation to the fronto-striatal circuits. CONCLUSION Increased understanding of the subcellular localization and unraveling of the signalosome concept of phosphodiesterases including its function and dysfunction in the fronto-striatal circuits will contribute to the design of new specific inhibitors and enhance the potential of phosphodiesterase inhibitors as therapeutics in fronto-striatal circuits.
Collapse
|
26
|
Raker VK, Becker C, Steinbrink K. The cAMP Pathway as Therapeutic Target in Autoimmune and Inflammatory Diseases. Front Immunol 2016; 7:123. [PMID: 27065076 PMCID: PMC4814577 DOI: 10.3389/fimmu.2016.00123] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/18/2016] [Indexed: 12/26/2022] Open
Abstract
Nucleotide signaling molecules contribute to the regulation of cellular pathways. In the immune system, cyclic adenosine monophosphate (cAMP) is well established as a potent regulator of innate and adaptive immune cell functions. Therapeutic strategies to interrupt or enhance cAMP generation or effects have immunoregulatory potential in autoimmune and inflammatory disorders. Here, we provide an overview of the cyclic AMP axis and its role as a regulator of immune functions and discuss the clinical and translational relevance of interventions with these processes.
Collapse
Affiliation(s)
- Verena Katharina Raker
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz , Mainz , Germany
| | - Christian Becker
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz , Mainz , Germany
| | - Kerstin Steinbrink
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz , Mainz , Germany
| |
Collapse
|
27
|
Corinaldesi C, Di Luigi L, Lenzi A, Crescioli C. Phosphodiesterase type 5 inhibitors: back and forward from cardiac indications. J Endocrinol Invest 2016; 39:143-51. [PMID: 26122487 PMCID: PMC4712255 DOI: 10.1007/s40618-015-0340-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/11/2015] [Indexed: 12/24/2022]
Abstract
PDE5 inhibitors (PDE5i) are widely known as treatment for erectile dysfunction (ED). This favorable action has emerged as a "side effect" from pioneering studies when PDE5i have been originally proposed as treatment for coronary artery disease (CAD). PDE5i showed marginal benefits for CAD treatment; although disappointing for that indication, they improved systemic and pulmonary vasodilation and ameliorated general endothelial function. Therefore, PDE5i have been approved and licensed also for pulmonary artery hypertension (PAH), besides ED. Nowadays, fine-tuned biomolecular mechanisms of PDE5i are well recognized to be beneficial onto myocardial contractility and geometry, to reduce tissue fibrosis, hypertrophy and apoptosis. PDE5i consistently exert benefits on heart failure, infarct, cardiomyopathy. The concept that PDE5i likely blunt Th1-driven inflammatory processes, which shift the homeostatic balance from health to disease, has emerged; PDE5i seem to decrease the release of active biomolecules from cells to tissues interested by inflammation. In this view, following clinical and basic research progresses, PDE5i can be undoubtedly "re-allocated" for cardiac indications and, hopefully, they could be approved as therapeutic tools to treat and prevent heart disease. This review aims to summarize PDE5i different clinical applications, from past to present and future, focusing on their potential power as treatment for cardiac diseases.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Cardiovascular Agents/pharmacokinetics
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/therapeutic use
- Cardiovascular Diseases/drug therapy
- Cardiovascular Diseases/immunology
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/physiopathology
- Coronary Artery Disease/drug therapy
- Coronary Artery Disease/immunology
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/physiopathology
- Cyclic Nucleotide Phosphodiesterases, Type 5/chemistry
- Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Evidence-Based Medicine
- Heart/drug effects
- Heart/physiopathology
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Myocardium/enzymology
- Myocardium/immunology
- Myocardium/metabolism
- Phosphodiesterase 5 Inhibitors/pharmacokinetics
- Phosphodiesterase 5 Inhibitors/pharmacology
- Phosphodiesterase 5 Inhibitors/therapeutic use
- Vasodilator Agents/pharmacokinetics
- Vasodilator Agents/pharmacology
- Vasodilator Agents/therapeutic use
Collapse
Affiliation(s)
- C Corinaldesi
- Department of Movement, Human and Health Sciences, Section of Health Science, Unit of Endocrinology, University of Rome "Foro Italico", Rome, Italy
| | - L Di Luigi
- Department of Movement, Human and Health Sciences, Section of Health Science, Unit of Endocrinology, University of Rome "Foro Italico", Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - C Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Science, Unit of Endocrinology, University of Rome "Foro Italico", Rome, Italy.
| |
Collapse
|
28
|
Feng Y, Cheng D, Zhang C, Li Y, Zhang Z, Wang J, Shi Y. Association of PDE4B Polymorphisms with Susceptibility to Schizophrenia: A Meta-Analysis of Case-Control Studies. PLoS One 2016; 11:e0147092. [PMID: 26756575 PMCID: PMC4710508 DOI: 10.1371/journal.pone.0147092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 12/26/2015] [Indexed: 11/29/2022] Open
Abstract
Background The PDE4B single nucleotide polymorphisms (SNPs) have been reported to be associated with schizophrenia risk. However, current findings are ambiguous or even conflicting. To better facilitate the understanding the genetic role played by PDE4B in susceptibility to schizophrenia, we collected currently available data and conducted this meta-analysis. Methods A comprehensive electronic literature searching of PubMed, Embase, Web of Science and Cochrane Library was performed. The association between PDE4B SNPs and schizophrenia was evaluated by odds ratios (ORs) and 95% confidence intervals (CIs) under allelic, dominant and recessive genetic models. The random effects model was utilized when high between-study heterogeneity (I2 > 50%) existed, otherwise the fixed effects model was used. Results Five studies comprising 2376 schizophrenia patients and 3093 controls were finally included for meta-analysis. The rs1040716 was statistically significantly associated with schizophrenia risk in Asian and Caucasian populations under dominant model (OR = 0.87, 95% CI: 0.76–0.99, P = 0.04). The rs2180335 was significantly related with schizophrenia risk in Asian populations under allelic (OR = 0.82, 95% CI: 0.72–0.93, P = 0.003) and dominant (OR = 0.75, 95% CI: 0.64–0.88, P < 0.001) models. A significant association was also observed between rs4320761 and schizophrenia in Asian populations under allelic model (OR = 0.87, 95% CI: 0.75–1.00, P = 0.048). In addition, a strong association tendency was found between rs6588190 and schizophrenia in Asian populations under allelic model (OR = 0.87, 95% CI: 0.76–1.00, P = 0.055). Conclusion This meta-analysis suggests that PDE4B SNPs are genetically associated with susceptibility to schizophrenia. However, due to limited sample size, more large-scale, multi-racial association studies are needed to further clarify the genetic association between various PDE4B variants and schizophrenia.
Collapse
Affiliation(s)
- Yanguo Feng
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Dejun Cheng
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Chaofeng Zhang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuchun Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhiying Zhang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Juan Wang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuzhong Shi
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- * E-mail:
| |
Collapse
|
29
|
An emerging role of cGMP in the treatment of schizophrenia: A review. Schizophr Res 2016; 170:226-31. [PMID: 26706197 DOI: 10.1016/j.schres.2015.11.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/12/2015] [Accepted: 11/16/2015] [Indexed: 01/08/2023]
Abstract
Schizophrenia is a progressive psychotic disorder with devastating effects on the broad aspects of human emotion, perception, thought, and psychosocial interactions. Although treatment with antipsychotic drugs, the mainstay in the treatment of schizophrenia, the large number of patients with schizophrenia respond poorly to the pharmacological and, the large number of patients with schizophrenia poorly respond to the pharmacological treatment. Although a variety of novel therapeutics have long been tested, to date, no drugs clinically efficacious for schizophrenia are available. The multiple lines of evidence strongly suggest that the modulation of cyclic guanosine monophosphate (cGMP) is a promising target in promoting the novel therapeutic strategies of schizophrenia beyond the "receptor-dependent" psychopharmacology. cGMP is modulated via regulating its synthesis by N-methyl-d-aspartate receptor (NMDAR) and nitric oxide (NO), which regulate guannylyl cyclase (GC), the enzyme producing cGMP. cGMP is also regulated by phosphodiesterase (PDE), the enzyme hydrolyzing cGMP. In this review, we critically evaluate the therapeutic potential of agents modulating cGMP activity by regulating cGMP synthesis including NMDAR enhancers, NO enhancers, NO inhibitors including minocycline with anti-inflammatory properties and PDE inhibitors in improving the negative, cognitive and positive symptoms of schizophrenia. We also discuss the possible mechanisms by which these agents produce therapeutic effects on schizophrenia including cGMP signaling pathways, oxidative stress, and neuroinflammation.
Collapse
|
30
|
Varnes JG, Geschwindner S, Holmquist CR, Forst J, Wang X, Dekker N, Scott CW, Tian G, Wood MW, Albert JS. Fragment-assisted hit investigation involving integrated HTS and fragment screening: Application to the identification of phosphodiesterase 10A (PDE10A) inhibitors. Bioorg Med Chem Lett 2015; 26:197-202. [PMID: 26597534 DOI: 10.1016/j.bmcl.2015.10.100] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/26/2015] [Accepted: 10/30/2015] [Indexed: 12/29/2022]
Abstract
Fragment-based drug design (FBDD) relies on direct elaboration of fragment hits and typically requires high resolution structural information to guide optimization. In fragment-assisted drug discovery (FADD), fragments provide information to guide selection and design but do not serve as starting points for elaboration. We describe FADD and high-throughput screening (HTS) campaign strategies conducted in parallel against PDE10A where fragment hit co-crystallography was not available. The fragment screen led to prioritized fragment hits (IC50's ∼500μM), which were used to generate a hypothetical core scaffold. Application of this scaffold as a filter to HTS output afforded a 4μM hit, which, after preparation of a small number of analogs, was elaborated into a 16nM lead. This approach highlights the strength of FADD, as fragment methods were applied despite the absence of co-crystallographical information to efficiently identify a lead compound for further optimization.
Collapse
Affiliation(s)
- Jeffrey G Varnes
- CNS Discovery Research, AstraZeneca Pharmaceuticals, 1800 Concord Pike, PO Box 15437, Wilmington, DE 19850-5437, USA.
| | | | - Christopher R Holmquist
- CNS Discovery Research, AstraZeneca Pharmaceuticals, 1800 Concord Pike, PO Box 15437, Wilmington, DE 19850-5437, USA
| | - Janet Forst
- CNS Discovery Research, AstraZeneca Pharmaceuticals, 1800 Concord Pike, PO Box 15437, Wilmington, DE 19850-5437, USA
| | - Xia Wang
- CNS Discovery Research, AstraZeneca Pharmaceuticals, 1800 Concord Pike, PO Box 15437, Wilmington, DE 19850-5437, USA
| | - Niek Dekker
- Discovery Sciences, AstraZeneca R&D, SE-431 83 Mölndal, Sweden
| | - Clay W Scott
- CNS Discovery Research, AstraZeneca Pharmaceuticals, 1800 Concord Pike, PO Box 15437, Wilmington, DE 19850-5437, USA
| | - Gaochao Tian
- CNS Discovery Research, AstraZeneca Pharmaceuticals, 1800 Concord Pike, PO Box 15437, Wilmington, DE 19850-5437, USA
| | - Michael W Wood
- CNS Discovery Research, AstraZeneca Pharmaceuticals, 1800 Concord Pike, PO Box 15437, Wilmington, DE 19850-5437, USA
| | - Jeffrey S Albert
- CNS Discovery Research, AstraZeneca Pharmaceuticals, 1800 Concord Pike, PO Box 15437, Wilmington, DE 19850-5437, USA.
| |
Collapse
|
31
|
Phosphodiesterase4D (PDE4D)--A risk factor for atrial fibrillation and stroke? J Neurol Sci 2015; 359:266-74. [PMID: 26671126 DOI: 10.1016/j.jns.2015.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 01/23/2023]
Abstract
Mutations in the gene encoding phosphodiesterase 4D (PDE4D) enzyme are associated with ischemic stroke; however the functional implications of such mutations are not well understood. PDE4D is part of a complex protein family modulating intracellular signalling by cyclic nucleotides. The PDE4 family includes subtypes A-D, all of which show unique intracellular, cellular and tissue distribution. PDE4D is the major subtype expressed in human atrial myocytes and involved in the pathophysiology of arrhythmias, such as atrial fibrillation. The PDE4D enzyme hydrolyses cyclic adenosine monophosphate (cAMP). Though diverging results are reported, several population based studies describe association of various PDE4D single nucleotide polymorphisms (SNP) with cardio-embolic stroke in particular. Functionally, a down regulation of PDE4D variants has been reported in stroke patients. The anti-inflammatory and vasodilator properties of PDE4 inhibitors make them suitable for treatment of stroke and cardiovascular disease. PDE4D has recently been suggested as factor in atrial fibrillation. This review summarizes the possible function of PDE4D in the brain, heart, and vasculature. Further, association of the described SNPs, in particular, with cardioembolic stroke, is reviewed. Current findings on the PDE4D mutations suggest functionality involves an increased cardiac risk factor as well as augmented risk of atrial fibrillation.
Collapse
|
32
|
Vlasceanu A, Jessing M, Kilburn JP. BN/CC isosterism in borazaronaphthalenes towards phosphodiesterase 10A (PDE10A) inhibitors. Bioorg Med Chem 2015; 23:4453-4461. [DOI: 10.1016/j.bmc.2015.06.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/01/2015] [Accepted: 06/05/2015] [Indexed: 10/23/2022]
|
33
|
The PDE10A inhibitor MP-10 and haloperidol produce distinct gene expression profiles in the striatum and influence cataleptic behavior in rodents. Neuropharmacology 2015; 99:256-63. [PMID: 26044638 DOI: 10.1016/j.neuropharm.2015.05.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 05/07/2015] [Accepted: 05/19/2015] [Indexed: 11/22/2022]
Abstract
Phosphodiesterase 10A (PDE10A) has garnered attention as a potential therapeutic target for schizophrenia due to its prominent striatal expression and ability to modulate striatal signaling. The present study used the selective PDE10A inhibitor MP-10 and the dopamine D2 antagonist haloperidol to compare effects of PDE10A inhibition and dopamine D2 blockade on striatopallidal (D2) and striatonigral (D1) pathway activation. Our studies confirmed that administration of MP-10 significantly elevates expression of the immediate early genes (IEG) c-fos, egr-1, and arc in rat striatum. Furthermore, we demonstrated that MP-10 induced egr-1 expression was distributed evenly between enkephalin-containing D2-neurons and substance P-containing D1-neurons. In contrast, haloperidol (3 mg/kg) selectively activated egr-1 expression in enkephalin neurons. Co-administration of MP-10 and haloperidol (0.5 mg/kg) increased IEG expression to a greater extent than either compound alone. Similarly, in a rat catalepsy assay, administration of haloperidol (0.5 mg/kg) or MP-10 (3-30 mg/kg) did not produce cataleptic behavior when dosed alone, but co-administration of haloperidol with MP-10 (3 and 10 mg/kg) induced cataleptic behaviors. Interestingly, co-administration of haloperidol with a high dose of MP-10 (30 mg/kg) failed to produce cataleptic behavior. These findings are important for understanding the neural circuits involved in catalepsy and suggest that the behavioral effects produced by PDE10A inhibitors may be influenced by concomitant medication and the level of PDE10A inhibition achieved by the dose of the inhibitor.
Collapse
|
34
|
Takano A, Stepanov V, Gulyás B, Nakao R, Amini N, Miura S, Kimura H, Taniguchi T, Halldin C. Evaluation of a novel PDE10A PET radioligand, [(11) C]T-773, in nonhuman primates: brain and whole body PET and brain autoradiography. Synapse 2015; 69:345-55. [PMID: 25892433 DOI: 10.1002/syn.21821] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 03/30/2015] [Accepted: 04/01/2015] [Indexed: 11/08/2022]
Abstract
Phosphodiesterase 10A (PDE10A) is considered to be a key target for the treatment of several neuropsychiatric diseases. The characteristics of [(11) C]T-773, a novel positron emission tomography (PET) radioligand with high binding affinity and selectivity for PDE10A, were evaluated in autoradiography and in nonhuman primate (NHP) PET. Brain PET measurements were performed under baseline conditions and after administration of a selective PDE10A inhibitor, MP-10. Total distribution volume (VT ) and binding potential (BPND ) were calculated using various kinetic models. Whole body PET measurements were performed to calculate the effective dose of [(11) C]T-773. Autoradiography studies in postmortem human and monkey brain sections showed high accumulation of [(11) C]T-773 in the striatum and substantia nigra which was blocked by MP-10. Brain PET showed high accumulation of [(11) C]T-773 in the striatum, and the data could be fitted using a two tissue compartment model. BPND was approximately 1.8 in the putamen when the cerebellum was used as the reference region. Approximately 70% of PDE10A binding was occupied by 1.8 mg/kg of MP-10. Whole body PET showed high accumulation of [(11) C]T-773 in the liver, kidney, heart, and brain in the initial phase. The radioligand was partly excreted via bile and the gastrointestinal tract, and partly excreted through the urinary tract. The calculated effective dose was 0.007 mSv/MBq. In conclusion, [(11) C]T-773 was demonstrated to be a promising PET radioligand for PDE10A with favorable brain kinetics. Dosimetry results support multiple PET measurements per person in human studies. Further research is required with [(11) C]T-773 in order to test the radioligand's potential clinical applications.
Collapse
Affiliation(s)
- Akihiro Takano
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Vladimir Stepanov
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Balázs Gulyás
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Ryuji Nakao
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Nahid Amini
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Shotaro Miura
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden.,CNS Drug Discovery Unit, Pharmaceutical Research Division, TAKEDA Pharmaceutical Company, Ltd., Fujisawa, Japan
| | - Haruhide Kimura
- CNS Drug Discovery Unit, Pharmaceutical Research Division, TAKEDA Pharmaceutical Company, Ltd., Fujisawa, Japan
| | - Takahiko Taniguchi
- CNS Drug Discovery Unit, Pharmaceutical Research Division, TAKEDA Pharmaceutical Company, Ltd., Fujisawa, Japan
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW The ketamine model has dominated drug discovery in schizophrenia over the past decade, supported by genetic and postmortem evidence implicating glutamatergic transmission. This review assesses recent successes and disappointments of glutamatergic agents and identifies promising new directions. RECENT FINDINGS Strategies focused on enhancing activity of the N-methyl D-aspartate (NMDA) receptor via direct agonists at the glycine site or by inhibition of glycine reuptake have produced modest and often inconsistent evidence of efficacy, as have approaches to reduce excessive glutamate release by lamotrigine or by mGluR2/3 agonists. Strategies targeting α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors have also met with only limited success. Newer approaches include selective allosteric modulation of NMDA receptor subunits and of mGluR5 receptors. In addition, intracellular pathways downstream of NMDA receptors may also provide new treatment targets, as exemplified by phosphodiesterase (PDE) inhibitors. SUMMARY Targeting glutamatergic transmission remains one of the most promising strategies in schizophrenia, particularly early in the course of illness, but therapeutic approaches may require greater specificity for receptor subtype type, illness phase, and individual biology in order to enhance efficacy and overcome problems with reproducibility of clinical results.
Collapse
|
36
|
Nallapati SB, Sreenivas BY, Bankala R, Parsa KVL, Sripelly S, Mukkanti K, Pal M. 1,2,3-Triazoles derived from olanzapine: their synthesis via an ultrasound assisted CuAAC method and evaluation as inhibitors of PDE4B. RSC Adv 2015. [DOI: 10.1039/c5ra20380e] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An ultrasound assisted CuAAC method afforded novel 1,2,3-triazoles derived from olanzapine as inhibitors of PDE4B.
Collapse
Affiliation(s)
- Suresh Babu Nallapati
- Dr Reddy's Institute of Life Sciences
- University of Hyderabad Campus
- Hyderabad-500046
- India
- Institute of Science and Technology
| | - B. Yogi Sreenivas
- Dr Reddy's Institute of Life Sciences
- University of Hyderabad Campus
- Hyderabad-500046
- India
| | - Ramudu Bankala
- Dr Reddy's Institute of Life Sciences
- University of Hyderabad Campus
- Hyderabad-500046
- India
| | - Kishore V. L. Parsa
- Dr Reddy's Institute of Life Sciences
- University of Hyderabad Campus
- Hyderabad-500046
- India
| | - Shivashankar Sripelly
- Dr Reddy's Institute of Life Sciences
- University of Hyderabad Campus
- Hyderabad-500046
- India
| | - K. Mukkanti
- Institute of Science and Technology
- JNT University Hyderabad
- Hyderabad-500085
- India
| | - Manojit Pal
- Dr Reddy's Institute of Life Sciences
- University of Hyderabad Campus
- Hyderabad-500046
- India
| |
Collapse
|
37
|
Rzasa RM, Frohn MJ, Andrews KL, Chmait S, Chen N, Clarine JG, Davis C, Eastwood HA, Horne DB, Hu E, Jones AD, Kaller MR, Kunz RK, Miller S, Monenschein H, Nguyen T, Pickrell AJ, Porter A, Reichelt A, Zhao X, Treanor JJ, Allen JR. Synthesis and preliminary biological evaluation of potent and selective 2-(3-alkoxy-1-azetidinyl) quinolines as novel PDE10A inhibitors with improved solubility. Bioorg Med Chem 2014; 22:6570-6585. [DOI: 10.1016/j.bmc.2014.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 10/01/2014] [Accepted: 10/12/2014] [Indexed: 01/13/2023]
|
38
|
Piel M, Vernaleken I, Rösch F. Positron Emission Tomography in CNS Drug Discovery and Drug Monitoring. J Med Chem 2014; 57:9232-58. [DOI: 10.1021/jm5001858] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Markus Piel
- Institute
of Nuclear Chemistry, Johannes Gutenberg-University, Fritz-Strassmann-Weg 2, D-55128 Mainz, Germany
| | - Ingo Vernaleken
- Department
of Psychiatry, Psychotherapy, and Psychosomatics, RWTH Aachen University, Pauwelsstraße 30, D-52074 Aachen, Germany
| | - Frank Rösch
- Institute
of Nuclear Chemistry, Johannes Gutenberg-University, Fritz-Strassmann-Weg 2, D-55128 Mainz, Germany
| |
Collapse
|
39
|
Megens AAHP, Hendrickx HMR, Mahieu MMA, Wellens ALY, de Boer P, Vanhoof G. PDE10A inhibitors stimulate or suppress motor behavior dependent on the relative activation state of the direct and indirect striatal output pathways. Pharmacol Res Perspect 2014; 2:e00057. [PMID: 25505601 PMCID: PMC4186443 DOI: 10.1002/prp2.57] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 05/09/2014] [Indexed: 11/09/2022] Open
Abstract
The enzyme phosphodiesterase 10A (PDE10A) regulates the activity of striatal, medium spiny neurons (MSNs), which are divided into a behaviorally stimulating, Gs-coupled D1 receptor-expressing “direct” pathway and a behaviorally suppressant, Gi-coupled D2 receptor-expressing “indirect” pathway. Activating both pathways, PDE10A inhibitors (PDE10AIs) combine functional characteristics of D2 antagonists and D1 agonists. While the effects of PDE10AIs on spontaneous and stimulated behavior have been extensively reported, the present study investigates their effects on suppressed behavior under various conditions of reduced dopaminergic neurotransmission: blockade of D1 receptors with SCH-23390, blockade of D2 receptors with haloperidol, or depletion of dopamine with RO-4-1284 or reserpine. In rats, PDE10AIs displayed relatively low cataleptic activity per se. After blocking D1 receptors, however, they induced pronounced catalepsy at low doses close to those required for inhibition of apomorphine-induced behavior; slightly higher doses resulted in behavioral stimulant effects, counteracting the catalepsy. PDE10AIs also counteracted catalepsy and related behaviors induced by D2 receptor blockade or dopamine depletion; catalepsy was replaced by behavioral stimulant effects under the latter but not the former condition. Similar interactions were observed at the level of locomotion in mice. At doses close to those inhibiting d-amphetamine-induced hyperlocomotion, PDE10AIs reversed hypolocomotion induced by D1 receptor blockade or dopamine depletion but not hypolocomotion induced by D2 receptor blockade. It is concluded that PDE10AIs stimulate or inhibit motor behavior dependent on the relative activation state of the direct and indirect striatal output pathways.
Collapse
Affiliation(s)
- Anton A H P Megens
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Herman M R Hendrickx
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Michel M A Mahieu
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Annemie L Y Wellens
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Peter de Boer
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| | - Greet Vanhoof
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV Beerse, Belgium
| |
Collapse
|
40
|
Babu PV, Gorja DR, Meda CLT, Deora GS, Kolli SK, Parsa KV, Mukkanti K, Pal M. Synthesis of N-(3-arylprop-2-ynyl)substituted olanzapine derivatives as potential inhibitors of PDE4B. Tetrahedron Lett 2014. [DOI: 10.1016/j.tetlet.2014.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
41
|
Critical role of nitric oxide in the modulation of prepulse inhibition in Swiss mice. Psychopharmacology (Berl) 2014; 231:663-72. [PMID: 24101156 DOI: 10.1007/s00213-013-3277-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 09/02/2013] [Indexed: 01/13/2023]
Abstract
RATIONALE Nitric oxide (NO) modulates the dopamine uptake and release processes and appears to be implicated in dopamine-related pathologies, such as schizophrenia. However, it is unclear whether there is excess or deficient NO synthesis in schizophrenia pathophysiology. Analyses of the intracellular pathways downstream of NO system activation have identified the cyclic nucleotide cyclic guanosine monophosphate (cGMP) as a possible target for drug development. Defects in the sensorimotor gating of the neural mechanism underlying the integration and processing of sensory information have been detected across species through prepulse inhibition (PPI). OBJECTIVES The aim of this study was to investigate the effects of NO/cGMP increase on sensorimotor gating modulation during dopamine hyperfunction. METHODS Mice were treated with NO donors and subjected to the PPI test. Treatment with the NO donor sodium nitroprusside was preceded by pretreatment with a soluble guanylate cyclase (sGC) inhibitor. Additionally, the mice were treated with NO donors and phosphodiesterases inhibitors prior to amphetamine treatment. RESULTS Pretreatment with the NO donors enhanced the PPI response and attenuated the amphetamine-disruptive effects on the PPI. The sGC inhibitor did not modify the sodium nitroprusside effects. Additionally, the cGMP increase induced by a specific phosphodiesterase inhibitor did not modify the amphetamine-disruptive effect. CONCLUSIONS This study provides the first demonstration that an increase in NO can improve the PPI response and block the amphetamine-disruptive effects on the PPI response. Our data are consistent with recent clinical results. However, these effects do not appear to be related to an increase in cGMP levels, and further investigation is thus required.
Collapse
|
42
|
Megens AAHP, Hendrickx HMR, Hens KA, Fonteyn I, Langlois X, Lenaerts I, Somers MVF, de Boer P, Vanhoof G. Pharmacology of JNJ-42314415, a Centrally Active Phosphodiesterase 10A (PDE10A) Inhibitor: A Comparison of PDE10A Inhibitors with D2 Receptor Blockers as Potential Antipsychotic Drugs. J Pharmacol Exp Ther 2014; 349:138-54. [DOI: 10.1124/jpet.113.211904] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
43
|
Banerjee A, Narayana L, Raje FA, Pisal DV, Kadam PA, Gullapalli S, Kumar H, More SV, Bajpai M, Sangana RR, Jadhav S, Gudi GS, Khairatkar-Joshi N, Merugu RR, Voleti SR, Gharat LA. Discovery of benzo[d]imidazo[5,1-b]thiazole as a new class of phosphodiesterase 10A inhibitors. Bioorg Med Chem Lett 2013; 23:6747-54. [DOI: 10.1016/j.bmcl.2013.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 10/04/2013] [Accepted: 10/17/2013] [Indexed: 01/19/2023]
|
44
|
Kilburn JP, Kehler J, Langgård M, Erichsen MN, Leth-Petersen S, Larsen M, Christoffersen CT, Nielsen J. N-Methylanilide and N-methylbenzamide derivatives as phosphodiesterase 10A (PDE10A) inhibitors. Bioorg Med Chem 2013; 21:6053-62. [DOI: 10.1016/j.bmc.2013.07.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 07/09/2013] [Accepted: 07/12/2013] [Indexed: 10/26/2022]
|
45
|
Regulation of hippocampal cGMP levels as a candidate to treat cognitive deficits in Huntington's disease. PLoS One 2013; 8:e73664. [PMID: 24040016 PMCID: PMC3764028 DOI: 10.1371/journal.pone.0073664] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/19/2013] [Indexed: 01/05/2023] Open
Abstract
Huntington’s disease (HD) patients and mouse models show learning and memory impairment associated with hippocampal dysfunction. The neuronal nitric oxide synthase/3',5'-cyclic guanosine monophosphate (nNOS/cGMP) pathway is implicated in synaptic plasticity, and in learning and memory processes. Here, we examined the nNOS/cGMP pathway in the hippocampus of HD mice to determine whether it can be a good therapeutic target for cognitive improvement in HD. We analyzed hippocampal nNOS and phosphodiesterase (PDE) 5 and 9 levels in R6/1 mice, and cGMP levels in the hippocampus of R6/1, R6/2 and HdhQ7/Q111 mice, and of HD patients. We also investigated whether sildenafil, a PDE5 inhibitor, could improve cognitive deficits in R6/1 mice. We found that hippocampal cGMP levels were 3-fold lower in 12-week-old R6/1 mice, when they show deficits in object recognition memory and in passive avoidance learning. Consistent with hippocampal cGMP levels, nNOS levels were down-regulated, while there were no changes in the levels of PDE5 and PDE9 in R6/1 mice. A single intraperitoneal injection of sildenafil (3 mg/Kg) immediately after training increased cGMP levels, and improved memory in R6/1 mice, as assessed by using the novel object recognition and the passive avoidance test. Importantly, cGMP levels were also reduced in R6/2 mouse and human HD hippocampus. Therefore, the regulation of hippocampal cGMP levels can be a suitable treatment for cognitive impairment in HD.
Collapse
|
46
|
PDE2 and PDE10, but not PDE5, inhibition affect basic auditory information processing in rats. Behav Brain Res 2013; 250:251-6. [DOI: 10.1016/j.bbr.2013.05.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/06/2013] [Accepted: 05/07/2013] [Indexed: 01/08/2023]
|
47
|
Giralt A, Saavedra A, Carretón O, Arumí H, Tyebji S, Alberch J, Pérez-Navarro E. PDE10 inhibition increases GluA1 and CREB phosphorylation and improves spatial and recognition memories in a Huntington's disease mouse model. Hippocampus 2013; 23:684-95. [PMID: 23576401 DOI: 10.1002/hipo.22128] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2013] [Indexed: 12/20/2022]
Abstract
Huntington's disease (HD) causes motor disturbances, preceded by cognitive impairment, in patients and mouse models. We showed that increased hippocampal cAMP-dependent protein kinase (PKA) signaling disrupts recognition and spatial memories in R6 HD mouse models. However, unchanged levels of hippocampal phosphorylated (p) cAMP-responsive element-binding protein (CREB) suggested unaltered nuclear PKA activity in R6 mice. Here, we extend this finding by showing that nuclear pPKA catalytic subunit (Thr197) and pPKA substrate levels were unaltered in the hippocampus of R6/1 mice. Phosphodiesterases (PDEs) play an important role in the regulation of PKA activity. PDE10A, a cAMP/cGMP dual-substrate PDE, was reported to be restricted to the nuclear region in nonstriatal neurons. Using cell fractionation we confirmed that PDE10A was enriched in nuclear fractions, both in wild-type and R6/1 mice hippocampus, without differences in its levels or intracellular distribution between genotypes. We next investigated whether inhibition of PDE10 with papaverine could improve cognitive function in HD mice. Papaverine treatment improved spatial and object recognition memories in R6/1 mice, and significantly increased pGluA1 and pCREB levels in R6/1 mice hippocampus. Papaverine likely acted through the activation of the PKA pathway as the phosphorylation level of distinct cGMP-dependent kinase (cGK) substrates was not modified in either genotype. Moreover, hippocampal cAMP, but not cGMP, levels were increased after acute papaverine injection. Our results show that inhibition of PDE10 improves cognition in R6 mice, at least in part through increased GluA1 and CREB phosphorylation. Thus, PDE10 might be a good therapeutic target to improve cognitive impairment in HD.
Collapse
Affiliation(s)
- Albert Giralt
- Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
48
|
Reneerkens OA, Rutten K, Bollen E, Hage T, Blokland A, Steinbusch HW, Prickaerts J. Inhibition of phoshodiesterase type 2 or type 10 reverses object memory deficits induced by scopolamine or MK-801. Behav Brain Res 2013; 236:16-22. [DOI: 10.1016/j.bbr.2012.08.019] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 08/10/2012] [Accepted: 08/14/2012] [Indexed: 12/31/2022]
|
49
|
Li J, Jin H, Zhou H, Rothfuss J, Tu Z. Synthesis and in vitro biological evaluation of pyrazole group-containing analogues for PDE10A. MEDCHEMCOMM 2013; 4:443-449. [PMID: 23585921 DOI: 10.1039/c2md20239e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Twenty eight new analogues were synthesized by optimizing the structure of MP-10 and their in vitro binding affinities towards PDE10A, PDE3A/B, and PDE4A/B were determined. Among these new analogues, 10a, 10b, 10d, 11a, 11b and 11d are very potent towards PDE10A and have IC50 values of 0.40 ± 0.02, 0.28 ± 0.06, 1.82 ± 0.25, 0.24 ± 0.05, 0.36 ± 0.03 and 1.78 ± 0.03 nM respectively; these six compounds displayed high selectivity for PDE10A versus PDE3A/3B/4A/4B. The promising compounds will be further validated in vivo to identify PDE10A imaging tracers.
Collapse
Affiliation(s)
- Junfeng Li
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA; ; Tel: +1-314-362-8487
| | | | | | | | | |
Collapse
|
50
|
Discovery of selective biaryl ethers as PDE10A inhibitors: improvement in potency and mitigation of Pgp-mediated efflux. Bioorg Med Chem Lett 2012; 22:7371-5. [PMID: 23149228 DOI: 10.1016/j.bmcl.2012.10.078] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 10/05/2012] [Accepted: 10/15/2012] [Indexed: 11/21/2022]
Abstract
We report the discovery of a novel series of biaryl ethers as potent and selective PDE10A inhibitors. Structure-activity studies improved the potency and decreased Pgp-mediated efflux found in the initial compound 4. X-ray crystallographic studies revealed two novel binding modes to the catalytic site of the PDE10A enzyme.
Collapse
|