Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
J Med Chem ; 67(10): 8309-8322, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38669059

ABSTRACT

Liver fibrosis is a common pathological feature of most chronic liver diseases with no effective drugs available. Phosphodiesterase 1 (PDE1), a subfamily of the PDE super enzyme, might work as a potent target for liver fibrosis by regulating the concentration of cAMP and cGMP. However, there are few PDE1 selective inhibitors, and none has been investigated for liver fibrosis treatment yet. Herein, compound AG-205/1186117 with the dihydropyrimidine scaffold was selected as the hit by virtual screening. A hit-to-lead structural modification led to a series of dihydropyrimidine derivatives. Lead 13h exhibited the IC50 of 10 nM against PDE1, high selectivity over other PDEs, as well as good safety properties. Administration of 13h exerted significant anti-liver fibrotic effects in bile duct ligation-induced fibrosis rats, which also prevented TGF-ß-induced myofibroblast differentiation in vitro, confirming that PDE1 could work as a potential target for liver fibrosis.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1 , Drug Design , Liver Cirrhosis , Phosphodiesterase Inhibitors , Pyrimidines , Animals , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Humans , Rats , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/therapeutic use , Phosphodiesterase Inhibitors/chemistry , Male , Structure-Activity Relationship , Rats, Sprague-Dawley , Molecular Docking Simulation , Molecular Structure
2.
Comput Biol Med ; 159: 106869, 2023 06.
Article in English | MEDLINE | ID: mdl-37071939

ABSTRACT

In recent years, the PDE1B enzyme has become a desirable drug target for the treatment of psychological and neurological disorders, particularly schizophrenia disorder, due to the expression of PDE1B in brain regions involved in volitional behaviour, learning and memory. Although several inhibitors of PDE1 have been identified using different methods, none of these inhibitors has reached the market yet. Thus, searching for novel PDE1B inhibitors is considered a major scientific challenge. In this study, pharmacophore-based screening, ensemble docking and molecular dynamics simulations have been performed to identify a lead inhibitor of PDE1B with a new chemical scaffold. Five PDE1B crystal structures have been utilised in the docking study to improve the possibility of identifying an active compound compared to the use of a single crystal structure. Finally, the structure-activity- relationship was studied, and the structure of the lead molecule was modified to design novel inhibitors with a high affinity for PDE1B. As a result, two novel compounds have been designed that exhibited a higher affinity to PDE1B compared to the lead compound and the other designed compounds.


Subject(s)
Molecular Dynamics Simulation , Pharmacophore , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Structure-Activity Relationship , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors
4.
Bioorg Chem ; 114: 105104, 2021 09.
Article in English | MEDLINE | ID: mdl-34186466

ABSTRACT

Phosphodiesterase-1 (PDE1) is a promising drug target closely related to central and peripheral diseases. With the assistance of molecular docking and dynamics simulations, we designed and synthesized a novel series of pyrazolopyrimidone derivatives as effective and metabolically stable inhibitors against PDE1. Most compounds have good inhibitory activities against PDE1 at the concentration of 20 nM. Compound 2j with the IC50 of 21 nM against PDE1B, shows good metabolic stability in the rat liver microsomes (RLM) (t1/2 of 28.5 min), indicating that compound 2j can be used as a tool to explore the molecular recognition mechanism between inhibitors and the target protein PDE1.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/pharmacology , Pyrimidinones/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Pyrimidinones/chemical synthesis , Pyrimidinones/chemistry , Structure-Activity Relationship
5.
Eur J Clin Invest ; 51(9): e13585, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34002387

ABSTRACT

INTRODUCTION: Phosphodiesterase (PDE) isoform inhibitors have mechanical and electrical effects on the heart. Inhibition of PDE-1 enzymes is a novel strategy for treating heart failure. However, the electrophysiological effects of PDE-1 inhibition on the heart remain unclear. This study explored the effects of PDE-1 inhibition using ITI-214 on electrical activity in the pulmonary vein (PV), the most common trigger of atrial fibrillation, and investigated the underlying ionic mechanisms. METHODS: Conventional microelectrodes or whole-cell patch clamps were employed to study the effects of ITI-214 (0.1-10 µM) on PV electrical activity, mechanical responses and ionic currents in isolated rabbit PV tissue specimens and isolated single PV cardiomyocytes. RESULTS: ITI-214 at 1 µM and 10 µM (but not 0.1 µM) significantly reduced PV spontaneous beating rate (10 ± 2% and 10 ± 3%, respectively) and PV diastolic tension (11 ± 3% and 17 ± 3%, respectively). ITI-24 (1 µM) significantly reduced late sodium current (INa-Late ), L-type calcium current (ICa-L ) and the reverse mode of the sodium-calcium exchanger (NCX), but it did not affect peak sodium currents. CONCLUSIONS: ITI-214 reduces PV spontaneous activity and PV diastolic tension by reducing INa-Late , ICa-L and NCX current. Considering its therapeutic potential in heart failure, targeting PDE-1 inhibition may provide a novel strategy for managing atrial arrhythmogenesis.


Subject(s)
Action Potentials/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Myocytes, Cardiac/drug effects , Phosphodiesterase Inhibitors/pharmacology , Pulmonary Veins/drug effects , Animals , Calcium/metabolism , Patch-Clamp Techniques , Pulmonary Veins/cytology , Rabbits
6.
Neuropharmacology ; 186: 108454, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33444639

ABSTRACT

Positive modulation of cAMP signalling by phosphodiesterase (PDE) inhibitors has recently been explored as a potential target for the reversal of cognitive and behavioural deficits implicating the corticoaccumbal circuit. Previous studies show that PDE type 1 isoform B (PDE1B) inhibition may improve memory function in rodent models; however, the contribution of PDE1B inhibition to impulsivity, attentional and motivational functions as well as its neurophysiological effects have not been investigated. To address this, we recorded single unit activity in medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) in Lister Hooded rats treated with the PDE1B inhibitor Lu AF64386 and tested in the 5-choice serial reaction time task (5-CSRTT). We also asked whether PDE1B inhibition modulates neurophysiological deficits produced by subchronic phencyclidine (PCP) treatment, a rat pharmacological model of schizophrenia. Lu AF64386 significantly affected behavioural parameters consistent with a reduction in goal-directed behaviour, however without affecting accuracy. Additionally, it reduced mPFC neuronal activity. Pre-treatment with PCP did not affect behavioural parameters, however it significantly disrupted overall neuronal firing while increasing phasic responses to reward-predicting cues and disrupting mPFC-NAc cross-talk. The latter two effects were reversed by Lu AF64386. These findings suggest PDE1B inhibition may be beneficial in disorders implicating a dysfunction of the mPFC-NAc network.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Goals , Phencyclidine/toxicity , Phosphodiesterase Inhibitors/therapeutic use , Prefrontal Cortex/enzymology , Schizophrenia/enzymology , Animals , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Disease Models, Animal , Female , Hallucinogens/toxicity , Phosphodiesterase Inhibitors/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiopathology , Rats , Schizophrenia/chemically induced , Schizophrenia/drug therapy
7.
J Cell Mol Med ; 24(24): 14514-14524, 2020 12.
Article in English | MEDLINE | ID: mdl-33169529

ABSTRACT

Hypoxic coronary vasospasm may lead to myocardial ischaemia and cardiac dysfunction. Inosine 3',5'-cyclic monophosphate (cIMP) is a putative second messenger to mediate this pathological process. Nevertheless, it remains unclear as to whether levels of cIMP can be regulated in living tissue such as coronary artery and if so, what is the consequence of this regulation on hypoxia-induced vasoconstriction. In the present study, we found that cIMP was a key determinant of hypoxia-induced constriction but not that of the subsequent relaxation response in porcine coronary arteries. Subsequently, coronary arteries were treated with various phosphodiesterase (PDE) inhibitors to identify PDE types that are capable of regulating cIMP levels. We found that inhibition of PDE1 and PDE5 substantially elevated cIMP content in endothelium-denuded coronary artery supplemented with exogenous purified cIMP. However, cGMP levels were far lower than their levels in intact coronary arteries and lower than cIMP levels measured in endothelium-denuded coronary arteries supplemented with exogenous cIMP. The increased cIMP levels induced by PDE1 or PDE5 inhibition further led to augmented hypoxic constriction without apparently affecting the relaxation response. In intact coronary artery, PDE1 or PDE5 inhibition up-regulated cIMP levels under hypoxic condition. Concomitantly, cGMP level increased to a comparable level. Nevertheless, the hypoxia-mediated constriction was enhanced in this situation that was largely compromised by an even stronger inhibition of PDEs. Taken together, these data suggest that cIMP levels in coronary arteries are regulated by PDE1 and PDE5, whose inhibition at a certain level leads to increased cIMP content and enhanced hypoxic constriction.


Subject(s)
Coronary Vessels/metabolism , Cyclic IMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Nitric Oxide/metabolism , Animals , Chromatography, High Pressure Liquid , Coronary Vessels/drug effects , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Hypoxia/metabolism , Metabolomics/methods , Phosphodiesterase 5 Inhibitors/pharmacology , Swine , Tandem Mass Spectrometry , Vasoconstriction
8.
Behav Brain Res ; 383: 112512, 2020 04 06.
Article in English | MEDLINE | ID: mdl-31991177

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease leading to cognitive and memory impairment. This study aimed at investigating the therapeutic and preserving effects of vinpocetine on amyloid beta (Aß)-induced rat model of AD. Sixty male adult Wistar rats were randomly divided into 6 groups (n = 10 per group) as follows: 1; control, 2; sham, 3; Aß, 4; pre-treatment (vinpocetine + Aß): oral gavage administration of vinpocetine at 4 mg/kg for 30 days followed by intracerebroventricular (ICV) injection of Aß, 5; treatment (Aß + vinpocetine): Aß ICV injection followed by vinpocetine administration for 30 days, 6; pre-treatment + treatment (vinpocetine + Aß + vinpocetine): vinpocetine administration for 30 days before and 30 days after AD induction. Following treatments, the animals' learning and memory were investigated using passive avoidance learning (PAL) task, Morris water maze (MWM), and novel object recognition (NOR) tests. The results demonstrated that Aß significantly enhanced escape latency and the distance traveled in the MWM, decreased step-through latency, and increased time spent in the dark compartment in PAL. Vinpocetine ameliorated the Aß-infused memory deficits in both MWM and PAL tests. Administration of vinpocetine in the Aß rats increased the discrimination index of the NOR test. It also significantly diminished the nitric oxide and malondialdehyde levels and restored the reduced glutathione (GSH) levels. Vinpocetine can improve memory and learning impairment following Aß infusion due to its different properties, including antioxidant effects, which indicates that vinpocetine administration can lead to the amelioration of cognitive dysfunction in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/toxicity , Memory/drug effects , Oxidative Stress/drug effects , Phosphodiesterase Inhibitors/pharmacology , Vinca Alkaloids/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Disease Models, Animal , Glutathione/drug effects , Glutathione/metabolism , Injections, Intraventricular , Learning/drug effects , Malondialdehyde/metabolism , Memory Disorders , Morris Water Maze Test , Nitrites/metabolism , Rats , Spatial Memory/drug effects
9.
Mol Cell Neurosci ; 102: 103449, 2020 01.
Article in English | MEDLINE | ID: mdl-31770590

ABSTRACT

A novel, potent, and highly specific inhibitor of calcium-calmodulin-dependent phosphodiesterases (PDE) of the PDE1 family, ITI-214, was used to investigate the role of PDE1 in inflammatory responses. ITI-214 dose-dependently suppressed lipopolysaccharide (LPS)-induced gene expression of pro-inflammatory cytokines in an immortalized murine microglial cell line, BV2 cells. RNA profiling (RNA-Seq) was used to analyze the impact of ITI-214 on the BV2 cell transcriptome in the absence and the presence of LPS. ITI-214 was found to regulate classes of genes that are involved in inflammation and cell migration responses to LPS exposure. The gene expression changes seen with ITI-214 treatment were distinct from those elicited by inhibitors of other PDEs with anti-inflammatory activity (e.g., a PDE4 inhibitor), indicating a distinct mechanism of action for PDE1. Functionally, ITI-214 inhibited ADP-induced migration of BV2 cells through a P2Y12-receptor-dependent pathway, possibly due to increases in the extent of cAMP and VASP phosphorylation downstream of receptor activation. Importantly, this effect was recapitulated in P2 rat microglial cells in vitro, indicating that these pathways are active in native microglial cells. These studies are the first to demonstrate that inhibition of PDE1 exerts anti-inflammatory effects through effects on microglia signaling pathways. The ability of PDE1 inhibitors to prevent or dampen excessive inflammatory responses of BV2 cells and microglia provides a basis for exploring their therapeutic utility in the treatment of neurodegenerative diseases associated with increased inflammation and microglia proliferation such as Parkinson's disease and Alzheimer's disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Microglia/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cell Line , Cell Movement , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cytokines/genetics , Cytokines/metabolism , Lipopolysaccharides/toxicity , Mice , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/physiology , Phosphoproteins/metabolism , Rats , Receptors, Purinergic P2Y12/metabolism , Signal Transduction
10.
Exp Parasitol ; 208: 107793, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31711973

ABSTRACT

Praziquantel (PZQ) is the sole drug used to treat schistosomiasis, and the probability of developing resistance is growing the longer it is relied upon, justifying the search for alternatives. Phosphodiesterases (PDEs), particularly the PDE4 family, have attracted considerable attention as drug targets, including in Schistosoma mansoni, and especially SmPDE4A. This study investigates the potential antischistosomal activity of human PDE4 and potent SmPDE4A inhibitor roflumilast, either alone or combined with PZQ. In vitro, roflumilast resulted in a significant, concentration-dependent reduction in egg production but not of worm viability. In vitro exposure to roflumilast in combination with a low concentration of PZQ was less effective than PZQ alone, pointing to antagonism. S. mansoni-infected mice treated with roflumilast showed significant reductions in worm burden (27%) as well as hepatic and intestinal egg burdens (~28%) two weeks post treatment. Scanning EM of worms isolated from roflumilast-treated and untreated mice did not reveal noticeable changes to their tegument. S. mansoni-infected mice treated with a fixed dosage of roflumilast and a variable dosage of PZQ resulted in a higher reduction in worm burden, reduced hepatic egg counts, absence of immature eggs and a marked increase in dead eggs, compared to PZQ alone. However, the combination resulted in increased animal mortality, probably attributable to pharmacodynamic interactions between the two drugs. Although this study marks the first report of in vivo antischistosomal potential by a PDE inhibitor, an important proof of concept, we conclude that the antischistosomal effects of roflumilast are insufficient to warrant further development.


Subject(s)
Aminopyridines/pharmacology , Anthelmintics/pharmacology , Benzamides/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Schistosoma mansoni/drug effects , Animals , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 1/drug effects , Cyclopropanes/pharmacology , Dose-Response Relationship, Drug , Female , Inhibitory Concentration 50 , Male , Mice , Microscopy, Electron, Scanning , Oviposition/drug effects , Praziquantel/pharmacology , Schistosoma mansoni/enzymology , Schistosoma mansoni/physiology , Schistosoma mansoni/ultrastructure
11.
Bioorg Med Chem Lett ; 30(2): 126472, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31859156

ABSTRACT

A new series of Vinpocetine derivatives were synthesized and evaluated for their inhibitory activity on PDE1A in vitro. Seven compounds with higher inhibitory activity were selected for surface plasmon resonance (SPR) binding experiments. Compared with Vinpocetine, these high potency compounds presented a higher binding affinity with PDE1A, which was consistent with inhibitory activity. After further screening, compounds 5, 7, 21, 34 and Vinpocetine were selected to examine the vasorelaxant effects on endothelium-intact rat thoracic aortic rings. The study suggested that the effects of compounds 7 and 21 were the most significant with the maximum value of 93.46 ±â€¯0.77% and 92.90 ±â€¯0.78% (n = 5) at a concentration of 100 µM respectively. Based on these studies, compounds 7 and 21 were considered for further development as hit compounds.


Subject(s)
Vasodilator Agents/chemical synthesis , Vinca Alkaloids/chemistry , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Kinetics , Rats , Structure-Activity Relationship , Surface Plasmon Resonance , Vasodilator Agents/metabolism , Vasodilator Agents/pharmacology , Vinca Alkaloids/metabolism , Vinca Alkaloids/pharmacology
12.
Cells ; 8(12)2019 11 20.
Article in English | MEDLINE | ID: mdl-31757003

ABSTRACT

Pharmacological activation of protein kinase A (PKA) reduces migration of arterial smooth muscle cells (ASMCs), including those isolated from human arteries (HASMCs). However, when individual migration-associated cellular events, including the polarization of cells in the direction of movement or rearrangements of the actin cytoskeleton, are studied in isolation, these individual events can be either promoted or inhibited in response to PKA activation. While pharmacological inhibition or deficiency of exchange protein activated by cAMP-1 (EPAC1) reduces the overall migration of ASMCs, the impact of EPAC1 inhibition or deficiency, or of its activation, on individual migration-related events has not been investigated. Herein, we report that EPAC1 facilitates the formation of leading-edge protrusions (LEPs) in HASMCs, a critical early event in the cell polarization that underpins their migration. Thus, RNAi-mediated silencing, or the selective pharmacological inhibition, of EPAC1 decreased the formation of LEPs by these cells. Furthermore, we show that the ability of EPAC1 to promote LEP formation by migrating HASMCs is regulated by a phosphodiesterase 1C (PDE1C)-regulated "pool" of intracellular HASMC cAMP but not by those regulated by the more abundant PDE3 or PDE4 activities. Overall, our data are consistent with a role for EPAC1 in regulating the formation of LEPs by polarized HASMCs and show that PDE1C-mediated cAMP hydrolysis controls this localized event.


Subject(s)
Aorta, Thoracic/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Myocytes, Smooth Muscle/metabolism , Signal Transduction , Aorta, Thoracic/drug effects , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Humans , Myocytes, Smooth Muscle/drug effects , Quinolines/pharmacology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects
13.
J Pharmacol Exp Ther ; 371(3): 692-702, 2019 12.
Article in English | MEDLINE | ID: mdl-31578257

ABSTRACT

In our drug discovery program, we identified a novel orally available and brain-penetrant phosphodiesterase (PDE) 1 inhibitor, 3-methyl-7-(tetrahydro-2H-pyran-4-yl)-2-{[trans-4-(trifluoromethyl)cyclohexyl]-methoxy}imidazo[5,1-f][1,2,4]triazin-4(3H)-one (DSR-141562). In the present study, we characterized the preclinical profile of DSR-141562. This compound has preferential selectivity for predominantly brain-expressed PDE1B over other PDE1 family members, and high selectivity for the PDE1 family over other PDE families and 65 other tested biologic targets. Oral administration of DSR-141562 at 10 mg/kg slightly elevated the cGMP concentration, and it potently enhanced the increase of cGMP induced by a dopamine D1 receptor agonist in mouse brains. The cGMP level in monkey cerebrospinal fluid was also elevated after treatment with DSR-141562 at 30 and 100 mg/kg and could be used as a translational biomarker. Since PDE1B is believed to regulate dopaminergic and glutamatergic signal transduction, we evaluated the effects of this compound using schizophrenia-related behavioral assays. DSR-141562 at 3-30 mg/kg potently inhibited methamphetamine-induced locomotor hyperactivity in rats, while it had only minimal effects on the spontaneous locomotor activity. Furthermore, DSR-141562 at 1-100 mg/kg did not induce any signs of catalepsy in rats. DSR-141562 at 0.3-3 mg/kg reversed social interaction and novel object recognition deficits induced by repeated treatment with an N-methyl-D-aspartate receptor antagonist, phencyclidine, in mice and rats, respectively. In common marmosets, DSR-141562 at 3 and 30 mg/kg improved the performance in object retrieval with detour tasks. These results suggest that DSR-141562 is a therapeutic candidate for positive, negative, and cognitive symptoms in schizophrenia. SIGNIFICANCE STATEMENT: This is the first paper showing that a phosphodiesterase 1 inhibitor is efficacious in animal models for positive and negative symptoms associated with schizophrenia. Furthermore, we demonstrated that this compound improved cognitive function in the common marmoset, a nonhuman primate.


Subject(s)
Cognition/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Imidazoles/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Schizophrenia/drug therapy , Triazines/pharmacology , Animals , Callithrix , Cyclic GMP/analysis , Cyclic GMP/cerebrospinal fluid , Disease Models, Animal , Female , Imidazoles/pharmacokinetics , Male , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Proto-Oncogene Proteins c-fos/genetics , Rats, Long-Evans , Receptors, Dopamine D1/physiology , Triazines/pharmacokinetics
14.
Neurochem Int ; 129: 104471, 2019 10.
Article in English | MEDLINE | ID: mdl-31121256

ABSTRACT

Phosphodiesterase (PDE) inhibition has been broadly investigated as a target for a wide variety of indications including central nervous system (CNS) disorders. Cyclic nucleotide (cNT) changes within associated tissues may serve as a biomarker of PDE inhibition. We recently developed robust sample harvesting and bioanalytical methods to quantify cNT levels in rodent brain and cerebrospinal fluid (CSF). Herein, we report on the application of those methods to study rodent species-specific and rodent brain region-specific cNT changes following individual or concomitant PDE inhibitor administration. Male Sprague Dawley (Crl:CD® [SD]) rats were dosed subcutaneously (sc) with a PDE1B inhibitor (DNS-0056), a PDE2A inhibitor (PF-05180999), a PDE9A inhibitor (PF-4447943), and a PDE10A inhibitor (MP10), each at a single dose of 10 or 30 mg/kg, or concomitantly with all 4 inhibitors at 10 mg/kg each. Male Carworth Farms (Crl:CF1 ®[CF-1]) mice were dosed intraperitoneally (ip) with the four individual inhibitors at a single dose of 10 mg/kg or concomitantly with all 4 inhibitors at 10 mg/kg each. The doses studied are generally adequate for affecting measurable cNT levels in the tissues of interest and were thereby chosen for this investigation. Measured 3',5'-cyclic adenosine monophosphate (cAMP) changes were generally statistically insignificant in the brain, striatum and CSF after administration of the aforementioned PDE inhibitors. However, the levels of 3',5'-cyclic guanosine monophosphate (cGMP) increased in both rat and mouse striatum (2.2-, 2.1- and 1.7-fold and 6.4-, 2.8- and 1.7-fold, respectively) after PDE2A, 9A, and 10A inhibitor dosing. In all cases, the cNT changes followed the same trend in the brain, striatum and CSF after PDE inhibitor dosing and dose response was observed in rats. Concomitant treatment with PDE1B, PDE2A, PDE9A and PDE10A inhibitors resulted in a 4.4- and 36.7-fold increase of cGMP in rat and mouse striatum. The drug exposures after concomitant treatment were also higher than in the individual inhibitor-treated animals. cGMP enhancement observed could be due to synergistic effects, though an additive effect of the combined inhibitor concentrations may also contribute.


Subject(s)
Central Nervous System/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/drug effects , Animals , Brain/drug effects , Brain/metabolism , Central Nervous System/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Male , Mice , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/metabolism , Rats, Sprague-Dawley
15.
Life Sci ; 222: 245-254, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30858122

ABSTRACT

BACKGROUND: Phosphodiestrase (PDE) enzymes are suggested to play a leading role in fibrogenesis of liver where studies showed the possible implication of PDE 1 & 4 in liver injury proposing them as possible targets for treating liver fibrosis. AIM: The present study was designed to investigate, for the first time, the possible therapeutic effects of selective inhibitors of PDE-1 (vinpocetine) and PDE-4 (roflumilast) in liver fibrosis induced by diethylnitrosamine (DEN) in rats. MAIN METHODS: Rats were given DEN (100 mg/kg, i.p.) once weekly for 6 weeks to induce liver fibrosis. Vinpocetine (10 mg/kg/day) or roflumilast (0.5 mg/kg/day) was then orally administered for 2 weeks. KEY FINDINGS: Vinpocetine significantly suppressed the contents of hydroxyproline, transforming growth factor-beta 1 (TGF-ß1), nuclear factor-kappa B (NF-κB) whereas roflumilast normalized them. Moreover, tumor necrosis factor-alpha (TNF-α) content and protein expressions of toll-like receptor 4 (TLR4) and tissue inhibitor of metalloproteinase-1 (TIMP-1) were markedly decreased whereas cAMP response element binding (CREB) protein expression was significantly elevated by both treatments. Additionally, vinpocetine and roflumilast up-regulated the gene expression of bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) receptor where roflumilast showed better results. PDE1 and 4 activities were inhibited by vinpocetine and roflumilast, respectively. The superior results offered by roflumilast could be related to the higher cAMP level obtained relative to vinpocetine. SIGNIFICANCE: Our study manifested the up-regulation of PDE enzymes (1 & 4) in liver fibrosis and addressed the therapeutic role of vinpocetine and roflumilast as PDEIs through a cAMP-mediated TLR4 inflammatory and fibrogenic signaling pathways.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Liver Cirrhosis/metabolism , Phosphodiesterase 4 Inhibitors/therapeutic use , Toll-Like Receptor 4/metabolism , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use , Liver Cirrhosis/drug therapy , Male , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Vinca Alkaloids/pharmacology , Vinca Alkaloids/therapeutic use
16.
J Agric Food Chem ; 67(5): 1429-1436, 2019 Feb 06.
Article in English | MEDLINE | ID: mdl-30628451

ABSTRACT

The aim of this work was to determine bioavailability and in vivo calmodulin-dependent-phosphodiesterase (CaMPDE)-inhibitory activity of six flaxseed-protein-derived peptides (AGA, AKLMS, QIAK, RWIQ, QQAKQ, and KQLSTGC) after oral administration to Wistar rats. Initial experiments tested the cytotoxicity and cellular-transport potentials of the peptides using Caco-2 cells. The cytotoxicity assay indicated that none of the six peptides had an adverse effect on the proliferation and viability of the Caco-2 cells, whereas the transport assay confirmed peptide translocation across the cell membrane. However, only two of the peptides (AGA and RWIQ) were detected in the rat serum up to 90 min postgavage, with traces of RWIQ persisting in serum 1 week after oral gavage. The six peptides inhibited plasma activity of CaMPDE with AGA (34.63%), QIAK (36.66%), and KQLSTGC (34.21%) being the most effective 30 min after gavage. In contrast, only AGA maintained significant plasma-CaMPDE-activity inhibition (44.35%) after 60 min.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Flax/chemistry , Peptides/chemistry , Animals , Biological Availability , Biological Transport , Caco-2 Cells , Cell Proliferation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/adverse effects , Female , Flax/genetics , Humans , Male , Peptide Mapping , Peptides/administration & dosage , Peptides/adverse effects , Rats , Rats, Wistar
17.
Exp Mol Med ; 51(1): 1-15, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635550

ABSTRACT

Vinpocetine, a phosphodiesterase (PDE) type-1 inhibitor, increases cAMP and cGMP levels and is currently used for the management of cerebrovascular disorders, such as stroke, cerebral hemorrhage, and cognitive dysfunctions. In this study, we first determined that vinpocetine effectively suppressed adipogenesis and lipid accumulation. However, we questioned which molecular mechanism is involved because the role of PDE in adipogenesis is still controversial. Vinpocetine decreased adipogenic cell signaling, including the phosphorylation of ERK, AKT, JAK2, and STAT3, and adipokine secretion, including IL-6, IL-10, and IFN-α. Interestingly, vinpocetine increased the phosphorylation of HSL, suggesting the induction of the lipolysis pathway. Moreover, vinpocetine increased UCP1 expression via increasing cAMP and PKA phosphorylation. The administration of vinpocetine with a normal-chow diet (NFD) or a high-fat diet (HFD) in mice attenuated body weight gain in mice fed both the NFD and HFD. These effects were larger in the HFD-fed mice, without a difference in food intake. Vinpocetine drastically decreased fat weight and adipocyte cell sizes in gonadal and inguinal white adipose tissues and in the liver in both diet groups. Serum triacylglycerol levels and fasting blood glucose levels were reduced by vinpocetine treatment. This study suggested that vinpocetine prevents adipocyte differentiation through the inhibition of adipogenesis-associated cell signaling in the early stages of adipogenesis. Moreover, upregulating cAMP levels leads to an increase in lipolysis and UCP1 expression and then inhibits lipid accumulation. Therefore, we suggest that vinpocetine could be an effective agent for treating obesity, as well as improving cognition and cardiovascular function in older individuals.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Lipolysis/drug effects , Phosphodiesterase Inhibitors/pharmacology , Vinca Alkaloids/pharmacology , 3T3 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipokines/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , Uncoupling Protein 1/metabolism , Weight Gain/drug effects
18.
Circulation ; 138(18): 1974-1987, 2018 10 30.
Article in English | MEDLINE | ID: mdl-30030415

ABSTRACT

BACKGROUND: Phosphodiesterase type-1 (PDE1) hydrolyzes cAMP and cGMP and is constitutively expressed in the heart, although cardiac effects from its acute inhibition in vivo are largely unknown. Existing data are limited to rodents expressing mostly the cGMP-favoring PDE1A isoform. Human heart predominantly expresses PDE1C with balanced selectivity for cAMP and cGMP. Here, we determined the acute effects of PDE1 inhibition in PDE1C-expressing mammals, dogs, and rabbits, in normal and failing hearts, and explored its regulatory pathways. METHODS: Conscious dogs chronically instrumented for pressure-volume relations were studied before and after tachypacing-induced heart failure (HF). A selective PDE1 inhibitor (ITI-214) was administered orally or intravenously±dobutamine. Pressure-volume analysis in anesthetized rabbits tested the role of ß-adrenergic and adenosine receptor signaling on ITI-214 effects. Sarcomere and calcium dynamics were studied in rabbit left ventricular myocytes. RESULTS: In normal and HF dogs, ITI-214 increased load-independent contractility, improved relaxation, and reduced systemic arterial resistance, raising cardiac output without altering systolic blood pressure. Heart rate increased, but less so in HF dogs. ITI-214 effects were additive to ß-adrenergic receptor agonism (dobutamine). Dobutamine but not ITI-214 increased plasma cAMP. ITI-214 induced similar cardiovascular effects in rabbits, whereas mice displayed only mild vasodilation and no contractility effects. In rabbits, ß-adrenergic receptor blockade (esmolol) prevented ITI-214-mediated chronotropy, but inotropy and vasodilation remained unchanged. By contrast, adenosine A2B-receptor blockade (MRS-1754) suppressed ITI-214 cardiovascular effects. Adding fixed-rate atrial pacing did not alter the findings. ITI-214 alone did not affect sarcomere or whole-cell calcium dynamics, whereas ß-adrenergic receptor agonism (isoproterenol) or PDE3 inhibition (cilostamide) increased both. Unlike cilostamide, which further enhanced shortening and peak calcium when combined with isoproterenol, ITI-214 had no impact on these responses. Both PDE1 and PDE3 inhibitors increased shortening and accelerated calcium decay when combined with forskolin, yet only cilostamide increased calcium transients. CONCLUSIONS: PDE1 inhibition by ITI-214 in vivo confers acute inotropic, lusitropic, and arterial vasodilatory effects in PDE1C-expressing mammals with and without HF. The effects appear related to cAMP signaling that is different from that provided via ß-adrenergic receptors or PDE3 modulation. ITI-214, which has completed phase I trials, may provide a novel therapy for HF.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Myocytes, Cardiac/physiology , Animals , Calcium/metabolism , Cyclic AMP/blood , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 3/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Dobutamine/therapeutic use , Dogs , Heart Failure/drug therapy , Heart Failure/etiology , Heart Rate/drug effects , Hemodynamics/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Rabbits , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects
19.
Circulation ; 138(18): 1988-2002, 2018 10 30.
Article in English | MEDLINE | ID: mdl-29871977

ABSTRACT

BACKGROUND: cAMP plays a critical role in regulating cardiomyocyte survival. Various cAMP signaling pathways behave distinctly or in opposition. We have previously reported that activation of cAMP hydrolysis by cyclic nucleotide phosphodiesterase 1C (PDE1C) promotes cardiomyocytes death/apoptosis, yet the underlying molecular mechanism remains unknown. In this study, we aimed to identify the specific cAMP signaling pathway modulated by PDE1C and determine the mechanism by which Ca2+/calmodulin-stimulated PDE1C is activated. METHODS: To study cardiomyocyte death/apoptosis, we used both isolated mouse adult cardiomyocytes in vitro and doxorubicin-induced cardiotoxicity in vivo. We used a variety of pharmacological activators and inhibitors as well as genetically engineered molecular tools to manipulate the expression and activity of proteins of interest. RESULTS: We found that the protective effect of PDE1C inhibition/deficiency on Ang II or doxorubicin-induced cardiomyocyte death/apoptosis is dependent on cAMP-generating adenosine A2 receptors (A2Rs), suggesting that PDE1C's cAMP-hydrolyzing activity selectively modulates A2R-cAMP signaling in cardiomyocytes. In addition, we found that the effects of PDE1C activation on Ang II-mediated cAMP reduction and cardiomyocyte death are dependent on transient receptor potential-canonical (TRPC) channels, in particular TRPC3. We also observed synergistic protective effects on cardiomyocyte survival from the combination of A2R stimulation together with PDE1 or TRPC inhibition. Coimmunostaining and coimmunoprecipitation studies showed that PDE1C is localized in proximity with A2R and TRPC3 in the plasma membrane and perhaps T tubules. It is important to note that we found that doxorubicin-induced cardiac toxicity and dysfunction in mice are attenuated by the PDE1 inhibitor IC86340 or in PDE1C knockout mice, and this protective effect is significantly diminished by A2R antagonism. CONCLUSIONS: We have characterized a novel multiprotein complex comprised of A2R, PDE1C, and TRPC3, in which PDE1C is activated by TRPC3-derived Ca2+, thereby antagonizing A2R-cAMP signaling and promoting cardiomyocyte death/apoptosis. Targeting these molecules individually or in combination may represent a compelling therapeutic strategy for potentiating cardiomyocyte survival.


Subject(s)
Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Receptors, Adenosine A2/metabolism , TRPC Cation Channels/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Angiotensin II/toxicity , Animals , Apoptosis/drug effects , Calcium Signaling/drug effects , Cell Survival/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Doxorubicin/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Adenosine A2/chemistry , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics
20.
J Med Chem ; 61(10): 4635-4640, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29718668

ABSTRACT

We disclose the discovery and X-ray cocrystal data of potent, selective quinazoline inhibitors of PDE1. Inhibitor ( S)-3 readily attains free plasma concentrations above PDE1 IC50 values and has restricted brain access. The racemic compound 3 inhibits >75% of PDE hydrolytic activity in soluble samples of human myocardium, consistent with heightened PDE1 activity in this tissue. These compounds represent promising new tools to probe the value of PDE1 inhibition in the treatment of cardiovascular disease.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Drug Discovery , Myocardium/enzymology , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Quinazolines/chemistry , Cyclic AMP/metabolism , Humans , Models, Molecular , Molecular Structure , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...