Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 108
1.
J Neurosci ; 44(23)2024 Jun 05.
Article En | MEDLINE | ID: mdl-38637154

Cocaine use disorder is a significant public health issue without an effective pharmacological treatment. Successful treatments are hindered in part by an incomplete understanding of the molecular mechanisms that underlie long-lasting maladaptive plasticity and addiction-like behaviors. Here, we leverage a large RNA sequencing dataset to generate gene coexpression networks across six interconnected regions of the brain's reward circuitry from mice that underwent saline or cocaine self-administration. We identify phosphodiesterase 1b (Pde1b), a Ca2+/calmodulin-dependent enzyme that increases cAMP and cGMP hydrolysis, as a central hub gene within a nucleus accumbens (NAc) gene module that was bioinformatically associated with addiction-like behavior. Chronic cocaine exposure increases Pde1b expression in NAc D2 medium spiny neurons (MSNs) in male but not female mice. Viral-mediated Pde1b overexpression in NAc reduces cocaine self-administration in female rats but increases seeking in both sexes. In female mice, overexpressing Pde1b in D1 MSNs attenuates the locomotor response to cocaine, with the opposite effect in D2 MSNs. Overexpressing Pde1b in D1/D2 MSNs had no effect on the locomotor response to cocaine in male mice. At the electrophysiological level, Pde1b overexpression reduces sEPSC frequency in D1 MSNs and regulates the excitability of NAc MSNs. Lastly, Pde1b overexpression significantly reduced the number of differentially expressed genes (DEGs) in NAc following chronic cocaine, with discordant effects on gene transcription between sexes. Together, we identify novel gene modules across the brain's reward circuitry associated with addiction-like behavior and explore the role of Pde1b in regulating the molecular, cellular, and behavioral responses to cocaine.


Cocaine-Related Disorders , Cyclic Nucleotide Phosphodiesterases, Type 1 , Gene Regulatory Networks , Mice, Inbred C57BL , Nucleus Accumbens , Sex Characteristics , Animals , Male , Female , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Mice , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/metabolism , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/genetics , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Cocaine/pharmacology , Reward
2.
J Med Chem ; 67(10): 8309-8322, 2024 May 23.
Article En | MEDLINE | ID: mdl-38669059

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.


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
3.
Acta Biochim Biophys Sin (Shanghai) ; 54(3): 388-399, 2022 Mar 25.
Article En | MEDLINE | ID: mdl-35538034

Although the association of elevated homocysteine level with cardiac hypertrophy has been reported, the molecular mechanisms by which homocysteine induces cardiac hypertrophy remain inadequately understood. In this study we aim to uncover the roles of cyclic nucleotide phosphodiesterase 1 (PDE1) and endoplasmic reticulum (ER) stress and their relationship to advance the mechanistic understanding of homocysteine-induced cardiac cell hypertrophy. H9c2 cells and primary neonatal rat cardiomyocytes are exposed to homocysteine with or without ER stress inhibitor TUDCA or PDE1-specific inhibitor Lu AF58027, or transfected with siRNAs targeting PDE1 isoforms prior to homocysteine-exposure. Cell surface area is measured and ultrastructure is examined by transmission electron microscopy. Hypertrophic markers, PDE1 isoforms, and ER stress molecules are detected by q-PCR and western blot analysis. Intracellular cGMP and cAMP are measured by ELISA. The results show that homocysteine causes the enlargement of H9c2 cells, increases the expressions of hypertrophic markers ß-MHC and ANP, upregulates PDE1A and PDE1C, promotes the expressions of ER stress molecules, and causes ER dilatation and degranulation. TUDCA and Lu AF58027 downregulate ß-MHC and ANP, and alleviate cell enlargement. TUDCA decreases PDE1A and PDE1C levels. Silencing of PDE1C inhibits homocysteine-induced hypertrophy, whereas PDE1A knockdown has minor effect. Both cAMP and cGMP are decreased after homocysteine-exposure, while only cAMP is restored by Lu AF58027 and TUDCA. TUDCA and Lu AF58027 also inhibit cell enlargement, downregulate ANP, ß-MHC and PDE1C, and enhance cAMP level in homocysteine-exposed primary cardiomyocytes. ER stress mediates homocysteine-induced hypertrophy of cardiac cells via upregulating PDE1C expression Cyclic nucleotide, especially cAMP, is the downstream mediator of the ER stress-PDE1C signaling axis in homocysteine-induced cell hypertrophy.


Cyclic Nucleotide Phosphodiesterases, Type 1 , Endoplasmic Reticulum Stress , Homocysteine , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomegaly/metabolism , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Homocysteine/pharmacology , Myocytes, Cardiac/metabolism , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Rats , Taurochenodeoxycholic Acid/pharmacology
5.
Microbiologyopen ; 10(4): e1203, 2021 08.
Article En | MEDLINE | ID: mdl-34459556

Streptococcus mitis is a commensal bacterial species of the oral cavity, with the potential for opportunistic pathogenesis. For successful colonization, S. mitis must be able to adhere to surfaces of the oral cavity and survive and adapt to frequently changing environmental conditions. Cyclic-di-AMP (c-di-AMP) is a nucleotide second messenger, involved in the regulation of stress responses and biofilm formation in several bacterial species. Cyclic-di-AMP is produced by diadenylate cyclases and degraded by phosphodiesterases. We have previously shown that in S. mitis, one diadenylate cyclase (CdaA) and at least two phosphodiesterases (Pde1 and Pde2) regulate the intracellular concentration of c-di-AMP. In this study, we utilized S. mitis deletion mutants of cdaA, pde1, and pde2 to analyze the role of c-di-AMP signaling in various stress responses, biofilm formation, and adhesion to eukaryotic cells. Here, we demonstrate that the Δpde1 mutant displayed a tendency toward increased susceptibility to acetic acid at pH 4.0. Deletion of cdaA increases auto-aggregation of S. mitis but reduces biofilm formation on an abiotic surface. These phenotypes are more pronounced under acidic extracellular conditions. Inactivation of pde1 or pde2 reduced the tolerance to ciprofloxacin, and UV radiation and the Δpde1 mutant was more susceptible to Triton X-100, indicating a role for c-di-AMP signaling in responses to DNA damage and cell membrane perturbation. Finally, the Δpde2 mutant displayed a tendency toward a reduced ability to adhere to oral keratinocytes. Taken together, our results indicate an important role for c-di-AMP signaling in cellular processes important for colonization of the mouth.


Adaptation, Physiological/physiology , Bacterial Adhesion/physiology , Biofilms/growth & development , Cyclic AMP/metabolism , Second Messenger Systems/physiology , Streptococcus mitis/metabolism , Acetic Acid/pharmacology , Cell Line, Tumor , Ciprofloxacin/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Gene Deletion , Gene Expression Regulation, Bacterial/genetics , Humans , Keratinocytes/microbiology , Mouth/microbiology , Octoxynol/pharmacology , Phosphorus-Oxygen Lyases/genetics , Phosphorus-Oxygen Lyases/metabolism , Streptococcus mitis/growth & development , Stress, Physiological/physiology
6.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article En | MEDLINE | ID: mdl-34312235

Abdominal aortic aneurysm (AAA) is characterized by aorta dilation due to wall degeneration, which mostly occurs in elderly males. Vascular aging is implicated in degenerative vascular pathologies, including AAA. Cyclic nucleotide phosphodiesterases, by hydrolyzing cyclic nucleotides, play critical roles in regulating vascular structure remodeling and function. Cyclic nucleotide phosphodiesterase 1C (PDE1C) expression is induced in dedifferentiated and aging vascular smooth muscle cells (SMCs), while little is known about the role of PDE1C in aneurysm. We observed that PDE1C was not expressed in normal aorta but highly induced in SMC-like cells in human and murine AAA. In mouse AAA models induced by Angiotensin II or periaortic elastase, PDE1C deficiency significantly decreased AAA incidence, aortic dilation, and elastin degradation, which supported a causative role of PDE1C in AAA development in vivo. Pharmacological inhibition of PDE1C also significantly suppressed preestablished AAA. We showed that PDE1C depletion antagonized SMC senescence in vitro and/or in vivo, as assessed by multiple senescence biomarkers, including senescence-associated ß-galactosidase activity, γ-H2AX foci number, and p21 protein level. Interestingly, the role of PDE1C in SMC senescence in vitro and in vivo was dependent on Sirtuin 1 (SIRT1). Mechanistic studies further showed that cAMP derived from PDE1C inhibition stimulated SIRT1 activation, likely through a direct interaction between cAMP and SIRT1, which leads to subsequent up-regulation of SIRT1 expression. Our findings provide evidence that PDE1C elevation links SMC senescence to AAA development in both experimental animal models and human AAA, suggesting therapeutical significance of PDE1C as a potential target against aortic aneurysms.


Aortic Aneurysm, Abdominal/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Gene Expression Regulation, Enzymologic/physiology , Angiotensin II/toxicity , Animals , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Biomarkers , Cellular Senescence , Cyclic AMP , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Histones , Male , Mice , Mice, Knockout, ApoE , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Up-Regulation , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
7.
Bioorg Chem ; 114: 105104, 2021 09.
Article En | MEDLINE | ID: mdl-34186466

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.


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
8.
J Biol Chem ; 296: 100606, 2021.
Article En | MEDLINE | ID: mdl-33789162

In addition to maintaining cellular ER Ca2+ stores, store-operated Ca2+ entry (SOCE) regulates several Ca2+-sensitive cellular enzymes, including certain adenylyl cyclases (ADCYs), enzymes that synthesize the secondary messenger cyclic AMP (cAMP). Ca2+, acting with calmodulin, can also increase the activity of PDE1-family phosphodiesterases (PDEs), which cleave the phosphodiester bond of cAMP. Surprisingly, SOCE-regulated cAMP signaling has not been studied in cells expressing both Ca2+-sensitive enzymes. Here, we report that depletion of ER Ca2+ activates PDE1C in human arterial smooth muscle cells (HASMCs). Inhibiting the activation of PDE1C reduced the magnitude of both SOCE and subsequent Ca2+/calmodulin-mediated activation of ADCY8 in these cells. Because inhibiting or silencing Ca2+-insensitive PDEs had no such effects, these data identify PDE1C-mediated hydrolysis of cAMP as a novel and important link between SOCE and its activation of ADCY8. Functionally, we showed that PDE1C regulated the formation of leading-edge protrusions in HASMCs, a critical early event in cell migration. Indeed, we found that PDE1C populated the tips of newly forming leading-edge protrusions in polarized HASMCs, and co-localized with ADCY8, the Ca2+ release activated Ca2+ channel subunit, Orai1, the cAMP-effector, protein kinase A, and an A-kinase anchoring protein, AKAP79. Because this polarization could allow PDE1C to control cAMP signaling in a hyper-localized manner, we suggest that PDE1C-selective therapeutic agents could offer increased spatial specificity in HASMCs over agents that regulate cAMP globally in cells. Similarly, such agents could also prove useful in regulating crosstalk between Ca2+/cAMP signaling in other cells in which dysregulated migration contributes to human pathology, including certain cancers.


Arteries/cytology , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Muscle Cells/cytology , Signal Transduction , Biological Transport , Cell Movement , Gene Expression Regulation, Enzymologic , Humans , Kinetics
9.
Neuropharmacology ; 186: 108454, 2021 03 15.
Article En | MEDLINE | ID: mdl-33444639

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.


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
10.
J Cell Mol Med ; 24(24): 14514-14524, 2020 12.
Article En | MEDLINE | ID: mdl-33169529

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.


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
11.
Cancer Res ; 80(16): 3200-3214, 2020 08 15.
Article En | MEDLINE | ID: mdl-32606006

N 6-Methyladenosine (m6A) is the most abundant modification of mammalian mRNAs. RNA methylation fine tunes RNA stability and translation, altering cell fate. The fat mass- and obesity-associated protein (FTO) is an m6A demethylase with oncogenic properties in leukemia. Here, we show that FTO expression is suppressed in ovarian tumors and cancer stem cells (CSC). FTO inhibited the self-renewal of ovarian CSC and suppressed tumorigenesis in vivo, both of which required FTO demethylase activity. Integrative RNA sequencing and m6A mapping analysis revealed significant transcriptomic changes associated with FTO overexpression and m6A loss involving stem cell signaling, RNA transcription, and mRNA splicing pathways. By reducing m6A levels at the 3'UTR and the mRNA stability of two phosphodiesterase genes (PDE1C and PDE4B), FTO augmented second messenger 3', 5'-cyclic adenosine monophosphate (cAMP) signaling and suppressed stemness features of ovarian cancer cells. Our results reveal a previously unappreciated tumor suppressor function of FTO in ovarian CSC mediated through inhibition of cAMP signaling. SIGNIFICANCE: A new tumor suppressor function of the RNA demethylase FTO implicates m6A RNA modifications in the regulation of cyclic AMP signaling involved in stemness and tumor initiation.


Adenosine/analogs & derivatives , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Second Messenger Systems , Tumor Suppressor Proteins/metabolism , 3' Untranslated Regions/genetics , Adenosine/genetics , Adenosine/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Alternative Splicing , Animals , Ascites/metabolism , Carcinogenesis/metabolism , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Down-Regulation , Fallopian Tubes/metabolism , Female , Gene Knockdown Techniques , Heterografts , Humans , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/pathology , Ovary/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , Sequence Analysis, RNA , Spheroids, Cellular , Tissue Array Analysis , Transcriptome , Tumor Suppressor Proteins/genetics
12.
J Med Chem ; 63(14): 7867-7879, 2020 07 23.
Article En | MEDLINE | ID: mdl-32603117

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and devastating lung disease lacking effective therapy. To identify whether phosphodiesterase-1 (PDE1) inhibition could act as a novel target for the treatment of IPF, hit-to-lead structural optimizations were performed on the PDE9/PDE1 dual inhibitor (R)-C33, leading to compound 3m with an IC50 of 2.9 nM against PDE1C, excellent selectivity across PDE subfamilies, reasonable drug-like properties, and remarkable pharmacodynamic effects as an anti-IPF agent. Oral administration of compound 3m (10 mg/kg) exerted more significant anti-pulmonary fibrosis effects than pirfenidone (150 mg/kg) in a bleomycin-induced IPF rat model and prevented transforming growth factor-ß-induced fibroblast-to-myofibroblast conversion in vitro, indicating that PDE1 inhibition could serve as a novel target for the efficient treatment of IPF.


Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidinones/therapeutic use , Animals , Bleomycin , Cell Differentiation/drug effects , Drug Design , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Male , Molecular Structure , Myofibroblasts/drug effects , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/pharmacokinetics , Protein Binding , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Pyrimidinones/chemical synthesis , Pyrimidinones/metabolism , Pyrimidinones/pharmacokinetics , Rats, Sprague-Dawley , Structure-Activity Relationship , Thermodynamics
13.
Basic Clin Pharmacol Toxicol ; 127(2): 67-80, 2020 Aug.
Article En | MEDLINE | ID: mdl-31495057

Among ageing-related illnesses, cardiovascular disease (CVD) remains the leading cause of morbidity and mortality causing one-third of all deaths worldwide. Ageing evokes a number of functional, pharmacological and morphological changes in the vasculature, accompanied by a progressive failure of protective and homeostatic mechanisms, resulting in target organ damage. Impaired vasomotor, proliferation, migration, antithrombotic and anti-inflammatory function in both the endothelial and vascular smooth muscle cells are parts of the vascular ageing phenotype. The endothelium regulates these functions by the release of a wide variety of active molecules including endothelium-derived relaxing factors such as nitric oxide, prostacyclin (PGI2 ) and endothelium-derived hyperpolarization (EDH). During ageing, a functional decay of the nitric oxide pathway takes place. Nitric oxide signals to VSMC and other important cell types for vascular homeostasis through the second messenger cyclic guanosine monophosphate (cGMP). Maintenance of proper cGMP levels is an important goal in sustainment of proper vascular function during ageing. For this purpose, different components can be targeted in this signalling system, and among them, phosphodiesterase-1 (PDE1) and soluble guanylate cyclase (sGC) are crucial. This review focuses on the role of PDE1 and sGC in conditions that are relevant for vascular ageing.


Aging/metabolism , Cardiovascular Diseases/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Nitric Oxide/biosynthesis , Soluble Guanylyl Cyclase/metabolism , Humans , Signal Transduction
14.
Mol Cell Neurosci ; 102: 103449, 2020 01.
Article En | MEDLINE | ID: mdl-31770590

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.


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
15.
Bioorg Med Chem Lett ; 30(2): 126472, 2020 01 15.
Article En | MEDLINE | ID: mdl-31859156

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.


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
16.
Cells ; 8(12)2019 11 20.
Article En | MEDLINE | ID: mdl-31757003

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.


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
17.
Curr Biol ; 29(15): 2580-2585.e4, 2019 08 05.
Article En | MEDLINE | ID: mdl-31353185

Although UVA radiation (315-400 nm) represents 95% of the UV radiation reaching the earth's surface, surprisingly little is known about its effects on plants [1]. We show that in Arabidopsis, short-term exposure to UVA inhibits the opening of stomata, and this requires a reduction in the cytosolic level of cGMP. This process is independent of UVR8, the UVB receptor. A cGMP-activated phosphodiesterase (AtCN-PDE1) was responsible for the UVA-induced decrease in cGMP in Arabidopsis. AtCN-PDE1-like proteins form a clade within the large HD-domain/PDEase-like protein superfamily, but no eukaryotic members of this subfamily have been functionally characterized. These genes have been lost from the genomes of metazoans but are otherwise conserved as single-copy genes across the tree of life. In longer-term experiments, UVA radiation increased growth and decreased water-use efficiency. These experiments revealed that PDE1 is also a negative regulator of growth. As the PDE1 gene is ancient and not represented in animal lineages, it is likely that at least one element of cGMP signaling in plants has evolved differently to the system present in metazoans.


Arabidopsis Proteins/genetics , Arabidopsis/radiation effects , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Ultraviolet Rays , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Signal Transduction
18.
Neurochem Int ; 129: 104471, 2019 10.
Article En | MEDLINE | ID: mdl-31121256

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.


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
19.
Sci Adv ; 5(5): eaaw5870, 2019 05.
Article En | MEDLINE | ID: mdl-31131329

No current treatment targets cardiac proteotoxicity or can reduce mortality of heart failure (HF) with preserved ejection fraction (HFpEF). Selective degradation of misfolded proteins by the ubiquitin-proteasome system (UPS) is vital to the cell. Proteasome impairment contributes to HF. Activation of cAMP-dependent protein kinase (PKA) or cGMP-dependent protein kinase (PKG) facilitates proteasome functioning. Phosphodiesterase 1 (PDE1) hydrolyzes both cyclic nucleotides and accounts for most PDE activities in human myocardium. We report that PDE1 inhibition (IC86430) increases myocardial 26S proteasome activities and UPS proteolytic function in mice. Mice with CryABR120G-based proteinopathy develop HFpEF and show increased myocardial PDE1A expression. PDE1 inhibition markedly attenuates HFpEF, improves mouse survival, increases PKA-mediated proteasome phosphorylation, and reduces myocardial misfolded CryAB. Therefore, PDE1 inhibition induces PKA- and PKG-mediated promotion of proteasomal degradation of misfolded proteins and treats HFpEF caused by CryABR120G, representing a potentially new therapeutic strategy for HFpEF and heart disease with increased proteotoxic stress.


Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Heart Failure/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Folding , Proteostasis Deficiencies/metabolism , alpha-Crystallin B Chain/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Densitometry , Echocardiography , Female , Genotype , Heart Failure/physiopathology , Heart Ventricles/metabolism , Hemodynamics , Humans , Hydrolysis , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Myocytes, Cardiac/metabolism , Phosphorylation , Protein Denaturation , Proteostasis Deficiencies/physiopathology , Rats
20.
Life Sci ; 222: 245-254, 2019 Apr 01.
Article En | MEDLINE | ID: mdl-30858122

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.


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
...