Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 116(27): 13320-13329, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31209056

ABSTRACT

Cyclic AMP (cAMP) phosphodiesterase-4 (PDE4) enzymes degrade cAMP and underpin the compartmentalization of cAMP signaling through their targeting to particular protein complexes and intracellular locales. We describe the discovery and characterization of a small-molecule compound that allosterically activates PDE4 long isoforms. This PDE4-specific activator displays reversible, noncompetitive kinetics of activation (increased Vmax with unchanged Km), phenocopies the ability of protein kinase A (PKA) to activate PDE4 long isoforms endogenously, and requires a dimeric enzyme assembly, as adopted by long, but not by short (monomeric), PDE4 isoforms. Abnormally elevated levels of cAMP provide a critical driver of the underpinning molecular pathology of autosomal dominant polycystic kidney disease (ADPKD) by promoting cyst formation that, ultimately, culminates in renal failure. Using both animal and human cell models of ADPKD, including ADPKD patient-derived primary cell cultures, we demonstrate that treatment with the prototypical PDE4 activator compound lowers intracellular cAMP levels, restrains cAMP-mediated signaling events, and profoundly inhibits cyst formation. PDE4 activator compounds thus have potential as therapeutics for treating disease driven by elevated cAMP signaling as well as providing a tool for evaluating the action of long PDE4 isoforms in regulating cAMP-mediated cellular processes.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Dogs , Enzyme Activation/drug effects , Humans , Madin Darby Canine Kidney Cells , Phosphorylation , Polycystic Kidney Diseases/metabolism , Protein Isoforms
2.
Neurobiol Dis ; 159: 105485, 2021 11.
Article in English | MEDLINE | ID: mdl-34411704

ABSTRACT

Fragile X Syndrome (FXS) is caused by silencing the FMR1 gene which results in intellectual disability, hyperactivity, sensory hypersensitivity, autistic-like behavior, and susceptibility to seizures. This X-linked disorder is also associated with reduced cAMP levels in humans as well as animal models. We assessed the therapeutic and neurochemical effects of chronic administration of the phosphodiesterase-4D negative allosteric modulator, BPN14770, in a mouse model of FXS (Fmr1 KO). Groups of male Fmr1 KO mice and control littermates were treated with dietary BPN14770 commencing postnatal day 21. A dose-response effect was investigated. At 90 days of age, mice underwent behavior tests including open field, novel object recognition, three chambered sociability and social novelty tests, passive avoidance, and sleep duration analysis. These tests were followed by in vivo measurement of regional rates of cerebral protein synthesis (rCPS) with the autoradiographic L-[1-14C]leucine method. BPN14770 treatment had positive effects on the behavioral phenotype in Fmr1 KO mice. Some effects such as increased sleep duration and increased social behavior occurred in both genotypes. In the open field, the hyperactivity response in Fmr1 KO mice was ameliorated by BPN14770 treatment at low and intermediate doses. BPN14770 treatment tended to increase rCPS in a dose-dependent manner in WT mice, whereas in Fmr1 KO mice effects on rCPS were less apparent. Results indicate BPN14770 treatment improves some behavior in Fmr1 KO mice. Results also suggest a genotype difference in the regulation of translation via a cAMP-dependent pathway.


Subject(s)
Behavior, Animal , Cerebrum , Fragile X Syndrome , Phosphodiesterase 4 Inhibitors , Protein Biosynthesis , Sleep , Animals , Mice , Allosteric Regulation , Autoradiography , Behavior, Animal/drug effects , Cerebrum/drug effects , Cerebrum/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Mice, Knockout , Phosphodiesterase 4 Inhibitors/pharmacology , Protein Biosynthesis/drug effects , Sleep/drug effects , Social Behavior
3.
J Am Acad Dermatol ; 78(3 Suppl 1): S43-S52, 2018 03.
Article in English | MEDLINE | ID: mdl-29248522

ABSTRACT

Historically, drugs available for treating atopic dermatitis (AD) have been limited to topical corticosteroids and topical calcineurin inhibitors, with systemic immunosuppressants and phototherapy reserved for severe AD. Despite their efficacy and infrequent adverse events, phobia about the use of topical steroids and calcineurin inhibitors has limited their use. More targeted options with fewer systemic and cutaneous side effects are needed for treating AD. Phosphodiesterase 4 (PDE4) is involved in the regulation of proinflammatory cytokines via the degradation of cyclic adenosine monophosphate. PDE4 activity is increased in the inflammatory cells of patients with AD, leading to increased production of proinflammatory cytokines and chemokines. Targeting PDE4 reduces the production of these proinflammatory mediators in AD. Both topical and oral PDE4 inhibitors have a favorable safety profile. Crisaborole 2% ointment, a topical PDE4, is now US Food and Drug Administration-approved for children older than 2 years and adults in the treatment of AD. Crisaborole 2% ointment shows early and sustained improvement in disease severity and pruritus and other AD symptoms, with burning and/or stinging upon application as the only related adverse event. Other PDE4 inhibitors are currently in trials with promising efficacy and safety.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Molecular Targeted Therapy/methods , Phosphodiesterase 4 Inhibitors/therapeutic use , Administration, Topical , Adult , Boron Compounds/therapeutic use , Child , Clinical Trials, Phase III as Topic , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Dermatitis, Atopic/diagnosis , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Prognosis , Quality of Life , Randomized Controlled Trials as Topic , Severity of Illness Index , Treatment Outcome , United States , United States Food and Drug Administration
4.
J Biochem Mol Toxicol ; 32(12): e22223, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30273962

ABSTRACT

Recently, antiobesity studies using the method of inhibiting enzymatic activity of obesity-related enzymes as targets have received considerable attention. The aims of the current study were to investigate whether p-hydroxybenzyl alcohol (HBA), identified from Cudrania tricuspidata fruits with antiobesity effects, inhibits the activity of digestive and obesity-related enzymes and acts as an inhibitor against four target enzymes in kinetic studies. In vitro enzyme assays showed HBA at the highest concentration significantly reduced the enzymatic activity of four targets: pancreatic lipase (IC50 = 2.34-3.70 µM), α-glycosidase (IC50 = 9.08 µM), phosphodiesterase IV (IC50 = 4.99 µM), and citrate synthase (IC50 = 2.07 µM) enzymes. Based on the results of kinetic assays, the types of inhibition were investigated. Our findings indicate that HBA could have antiobesity efficacy, and it deserves further study.


Subject(s)
Anti-Obesity Agents/pharmacology , Benzyl Alcohols/pharmacology , Citrate (si)-Synthase/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Enzyme Inhibitors/pharmacology , Lipase/antagonists & inhibitors , Obesity/enzymology , alpha-Glucosidases/drug effects , Animals , Humans , Inhibitory Concentration 50 , Kinetics , Moraceae/chemistry , Plant Extracts/pharmacology , Swine
5.
Proc Natl Acad Sci U S A ; 112(7): 2023-8, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25646485

ABSTRACT

cAMP production and protein kinase A (PKA) are the most widely studied steps in ß-adrenergic receptor (ßAR) signaling in the heart; however, the multifunctional Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is also activated in response to ßAR stimulation and is involved in the regulation of cardiac excitation-contraction coupling. Its activity and expression are increased during cardiac hypertrophy, in heart failure, and under conditions that promote arrhythmias both in animal models and in the human heart, underscoring the clinical relevance of CaMKII in cardiac pathophysiology. Both CaMKII and PKA phosphorylate a number of protein targets critical for Ca(2+) handling and contraction with similar, but not always identical, functional consequences. How these two pathways communicate with each other remains incompletely understood, however. To maintain homeostasis, cyclic nucleotide levels are regulated by phosphodiesterases (PDEs), with PDE4s predominantly responsible for cAMP degradation in the rodent heart. Here we have reassessed the interaction between cAMP/PKA and Ca(2+)/CaMKII signaling. We demonstrate that CaMKII activity constrains basal and ßAR-activated cAMP levels. Moreover, we show that these effects are mediated, at least in part, by CaMKII regulation of PDE4D. This regulation establishes a negative feedback loop necessary to maintain cAMP/CaMKII homeostasis, revealing a previously unidentified function for PDE4D as a critical integrator of cAMP/PKA and Ca(2+)/CaMKII signaling.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Feedback , Signal Transduction , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Humans , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation
6.
Glia ; 64(10): 1698-709, 2016 10.
Article in English | MEDLINE | ID: mdl-27038323

ABSTRACT

The importance of microglia in immune homeostasis within the brain is undisputed. Their role in a diversity of neurological and psychiatric diseases as well as CNS injury is the subject of much investigation. Cyclic adenosine monophosphate (AMP) is a critical regulator of microglia homeostasis; as the predominant negative modulator of cyclic AMP signaling within microglia, phosphodiesterase 4 (PDE4) represents a promising target for modulating immune function. PDE4 expression is regulated by inflammation, and in turn, PDE4 inhibition can alter microglia reactivity. As the prototypic PDE4 inhibitor, rolipram, was tested clinically in the 1980s, drug discovery and clinical development of PDE4 inhibitors have been severely hampered by tolerability issues involving nausea and emesis. The two PDE4 inhibitors approved for peripheral inflammatory disorders (roflumilast and apremilast) lack brain penetration and are dose-limited by side effects making them unsuitable for modulating microglial function. Subtype selective inhibitors targeting PDE4B are of high interest given the critical role PDE4B plays in immune function versus the association of PDE4D with nausea and emesis. The challenges and requirements for successful development of a novel brain-penetrant PDE4B inhibitor are discussed in the context of early clinical development strategies. Furthermore, the challenges of monitoring the state of microglia in vivo are highlighted, including a description of the currently available tools and their limitations. Continued drug discovery efforts to identify safe and well-tolerated, brain-penetrant PDE4 inhibitors are a reflection of the confidence in the rationale for modulation of this target to produce meaningful therapeutic benefit in a wide range of neurological conditions and injury. GLIA 2016;64:1698-1709.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Microglia/drug effects , Microglia/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Encephalitis/drug therapy , Encephalitis/metabolism , Encephalitis/pathology , Humans
7.
Arch Pharm (Weinheim) ; 349(12): 889-903, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27869315

ABSTRACT

In the search for potential psychotropic agents, a new series of 3,7-dimethyl- and 1,3-dimethyl-8-alkoxypurine-2,6-dione derivatives of arylpiperazines, perhydroisoquinolines, or tetrahydroisoquinolines with flexible alkylene spacers (5-16 and 21-32) were synthesized and evaluated for 5-HT1A /5-HT7 receptor affinities as well as PDE4B1 and PDE10A inhibitory properties. The 1-(4-(4-(2-hydroxyphenyl)piperazin-1-yl)butyl)-3,7-dimethyl-8-propoxypurine-2,6-dione (16) and 7-(2-hydroxyphenyl)piperazinylalkyl-1,3-dimethyl-8-ethoxypurine-2,6-diones (31 and 32) as potent dual 5-HT1A /5-HT7 receptor ligands with antagonistic activity produced an antidepressant-like effect in the forced swim test in mice. This effect was similar to that produced by citalopram. All the tested compounds were stronger phosphodiesterase isoenzyme inhibitors than theophylline and theobromine. The most potent compounds, 15 and 16, were characterized by 51 and 52% inhibition, respectively, of PDE4B1 activity at a concentration of 10-5 M. Concerning the above findings, it may be assumed that the inhibition of PDE4B1 may impact on the signal strength and specificity resulting from antagonism toward the 5-HT1 and 5-HT7 receptors, especially in the case of compounds 15 and 16. This dual receptor and enzyme binding mode was analyzed and explained via molecular modeling studies.


Subject(s)
Antidepressive Agents/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Piperazines/chemical synthesis , Piperazines/pharmacology , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Animals , Antidepressive Agents/chemical synthesis , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Immobility Response, Tonic/drug effects , Isoenzymes/antagonists & inhibitors , Isoquinolines/chemical synthesis , Isoquinolines/pharmacology , Male , Mice , Models, Molecular , Phosphodiesterase Inhibitors/chemical synthesis , Phosphoric Diester Hydrolases/drug effects , Receptors, Serotonin/drug effects , Serotonin Antagonists/chemical synthesis , Serotonin Receptor Agonists/chemical synthesis , Structure-Activity Relationship , Theobromine/pharmacology , Theophylline/pharmacology
8.
Allergy ; 70(12): 1622-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26335809

ABSTRACT

BACKGROUND: Asthma is a heterogenetic disorder characterized by chronic inflammation with variable airflow obstruction and airway hyper-responsiveness. As the most potent and popular bronchodilators, ß2 adrenergic receptor (ß2 AR) agonists bind to the ß2 ARs that are coupled via a stimulatory G protein to adenylyl cyclase, thereby improving cAMP accumulation and resulting in airway smooth muscle relaxation. We previously demonstrated arctigenin had a synergistic function with the ß2 AR agonist, but the target for this remained elusive. METHOD: Chemical proteomics capturing was used to enrich and uncover the target of arctigenin in human bronchial smooth muscle cells, and reverse docking and molecular dynamic stimulation were performed to evaluate the binding of arctigenin and its target. In vitro enzyme activities and protein levels were demonstrated with special kits and Western blotting. Finally, guinea pig tracheal muscle segregation and ex vivo function were analysed. RESULTS: Arctigenin bound to PDK1 with an ideal binding free energy -25.45 kcal/mol and inhibited PDK1 kinase activity without changing its protein level. Additionally, arctigenin reduced PKB/Akt-induced phosphorylation of PDE4D, which was first identified in this study. Attenuation of PDE4D resulted in cAMP accumulation in human bronchial smooth muscle. The inhibition of PDK1 showed a synergistic function with ß2 AR agonists and relaxed the constriction of segregated guinea pig tracheal muscle. CONCLUSIONS: The PDK1/Akt/PDE4D axis serves as a novel asthma target, which may benefit airflow obstruction.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/metabolism , Bronchi/drug effects , Furans/pharmacology , Lignans/pharmacology , Signal Transduction/drug effects , 3-Phosphoinositide-Dependent Protein Kinases/drug effects , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Blotting, Western , Bronchi/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Drug Synergism , Guinea Pigs , Humans , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism
9.
Biochem Biophys Res Commun ; 450(4): 1560-7, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-25019986

ABSTRACT

Acute lung injury (ALI), acute respiratory distress syndrome (ARDS), is actually involved in an ongoing and uncontrolled inflammatory response in lung tissues. Although extensive studies suggested that phospodiesterase type 4B (PDE4B) may be related to inflammation, the underlying cell biological mechanism of ALI remains unclear. To further investigate the mechanism how PDE4B take part in inflammatory response and the maintenance of vascular integrity, we established the experimental model of ALI in vitro and in vivo. In vitro, we found that Cilomilast, Diazepam and PDE4B knockout could potently inhibit the LPS-induced NF-κB activation and inflammatory response in multiple cell types, including lung epithelial cells (A549), pulmonary microvascular endothelial cells (PMVECs) and vascular smooth muscle cells (VSMCs). Besides, PDE4B deletion attenuated the LPS-induced ROS generation. In vivo, PDE4B deletion could attenuate the lung water content, histological signs of pulmonary injury and elevate the ratio of partial pressure of arterial O2 to fraction of inspired O2 (PaO2/FIO2 ratio). Additionally, PDE4B deletion reduced LPS-induced vascular permeability. Collectively, our results strongly indicates that PDE4B is a valid target for anti-ALI.


Subject(s)
Acute Lung Injury/chemically induced , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Lipopolysaccharides/toxicity , Phosphodiesterase 4 Inhibitors/pharmacology , Pneumonia/prevention & control , Acute Lung Injury/enzymology , Acute Lung Injury/physiopathology , Animals , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Male , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley
10.
J Pharmacol Exp Ther ; 350(1): 153-63, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24784567

ABSTRACT

Small molecule phosphodiesterase (PDE) 4 inhibitors have long been known to show therapeutic benefit in various preclinical models of psychiatric and neurologic diseases because of their ability to elevate cAMP in various cell types of the central nervous system. Despite the registration of the first PDE4 inhibitor, roflumilast, for the treatment of chronic obstructive pulmonary disease, the therapeutic potential of PDE4 inhibitors in neurologic diseases has never been fulfilled in the clinic due to severe dose-limiting side effects such as nausea and vomiting. In this study, we describe the detailed pharmacological characterization of GSK356278 [5-(5-((2,4-dimethylthiazol-5-yl)methyl)-1,3,4-oxadiazol-2-yl)-1-ethyl-N-(tetrahydro-2H-pyran-4-yl)-1H-pyrazolo[3,4-b]pyridin-4-amine], a potent, selective, and brain-penetrant PDE4 inhibitor that shows a superior therapeutic index to both rolipram and roflumilast in various preclinical species and has potential for further development in the clinic for the treatment of psychiatric and neurologic diseases. GSK356278 inhibited PDE4B enzyme activity with a pIC50 of 8.8 and bound to the high-affinity rolipram binding site with a pIC50 of 8.6. In preclinical models, the therapeutic index as defined in a rodent lung inflammation model versus rat pica feeding was >150 compared with 0.5 and 6.4 for rolipram and roflumilast, respectively. In a model of anxiety in common marmosets, the therapeutic index for GSK356278 was >10 versus <1 for rolipram. We also demonstrate that GSK356278 enhances performance in a model of executive function in cynomolgus macaques with no adverse effects, a therapeutic profile that supports further evaluation of GSK356278 in a clinical setting.


Subject(s)
Cerebral Cortex/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Nootropic Agents/pharmacology , Oxadiazoles/pharmacology , Phosphodiesterase 4 Inhibitors/adverse effects , Phosphodiesterase 4 Inhibitors/pharmacology , Thiazoles/pharmacology , Aminopyridines/pharmacology , Animals , Anti-Anxiety Agents/adverse effects , Anti-Anxiety Agents/pharmacokinetics , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Behavior, Animal/drug effects , Benzamides/pharmacology , Callithrix , Cerebral Cortex/drug effects , Cyclopropanes/pharmacology , Drug Evaluation, Preclinical , Ferrets , Inflammation/chemically induced , Inflammation/drug therapy , Isoenzymes/antagonists & inhibitors , Macaca fascicularis , Male , Nootropic Agents/adverse effects , Nootropic Agents/pharmacokinetics , Nootropic Agents/therapeutic use , Oxadiazoles/adverse effects , Oxadiazoles/pharmacokinetics , Oxadiazoles/therapeutic use , Phosphodiesterase 4 Inhibitors/pharmacokinetics , Pica/drug therapy , Rats , Rolipram/pharmacology , Thiazoles/adverse effects , Thiazoles/pharmacokinetics , Thiazoles/therapeutic use
11.
J Nat Prod ; 77(8): 1928-36, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25075977

ABSTRACT

Ten new prostaglandin derivatives (PGs), sarcoehrendins A-J (1-10), together with five known analogues (11-15) were isolated from the soft coral Sarcophyton ehrenbergi. Compounds 4-8 represented the first examples of PGs featuring an 18-ketone group. The structures including the absolute configurations were elucidated on the basis of spectroscopic analysis and chemical evidence. All of the isolates and six synthetic analogues (3a, 3b, 4a, and 11a-11c) were screened for inhibitory activity against phosphodiesterase-4 (PDE4), which is a drug target for the treatment of asthma and chronic obstructive pulmonary disease. Compounds 2, 10, 11a, 11b, and 13-15 exhibited inhibition with IC50 values less than 10 µM, and compound 15 (IC50 = 1.4 µM) showed comparable activity to the positive control rolipram (IC50 = 0.60 µM). The active natural PGs (2, 10, and 13-15) represent the first examples of PDE4 inhibitors without an aromatic moiety, and a preliminary structure-activity relationship is also proposed.


Subject(s)
Anthozoa/chemistry , Phosphodiesterase 4 Inhibitors/isolation & purification , Phosphodiesterase 4 Inhibitors/pharmacology , Prostaglandins/isolation & purification , Prostaglandins/pharmacology , Animals , China , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Phosphodiesterase 4 Inhibitors/chemistry , Prostaglandins/chemistry , Rolipram/pharmacology , Structure-Activity Relationship
12.
Am J Respir Crit Care Med ; 188(3): 271-8, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23656508

ABSTRACT

Phosphodiesterases (PDEs) are a superfamily of enzymes that catalyze the breakdown of cAMP and/or cyclic guanosine monophosphate (GMP) to their inactive form. PDE4 is the main selective cAMP-metabolizing enzyme in inflammatory and immune cells. Because PDE4 is highly expressed in leukocytes and other inflammatory cells involved in the pathogenesis of inflammatory lung diseases, such as asthma and chronic obstructive pulmonary disease (COPD), inhibition of PDE4 has been predicted to have an antiinflammatory effect and thus therapeutic efficacy. The limited and inconsistent efficacy and side effects of the early compounds made their further development less desirable in asthma, given the excellent efficacy/tolerability ratio of inhaled steroids. The lack of effective antiinflammatory drug treatment for COPD has thus shifted the interest in development toward COPD. Roflumilast, the only PDE4 inhibitor that has reached the market because of the good efficacy/tolerability ratio, is recommended for patients with COPD with severe airflow limitation, symptoms of chronic bronchitis, and a history of exacerbations, whose disease is not adequately controlled by long-acting bronchodilators. Albeit safe, it maintains significant side effects (diarrhea, nausea, weight loss) that make it intolerable in some patients. Future developments of PDE4 inhibitors include extended indications of roflumilast (1) in patients with COPD, and (2) in other respiratory (e.g., asthma) and nonrespiratory chronic inflammatory/metabolic conditions (e.g., diabetes), as well as (3) the development of new molecules with PDE4 inhibitory properties with an improved efficacy/tolerability profile.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Lung Diseases/drug therapy , Phosphodiesterase 4 Inhibitors/therapeutic use , Cyclic Nucleotide Phosphodiesterases, Type 4/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 4/blood , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Humans , Leukocytes/enzymology , Lung Diseases/enzymology
13.
Mar Drugs ; 12(2): 672-81, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24473168

ABSTRACT

Six new tetraprenylated alkaloids, designated as malonganenones L-Q (1-6), were isolated from the gorgonian Echinogorgia pseudossapo, collected in Daya Bay of Guangdong Province, China. The structures of 1-6 featuring a methyl group at N-3 and a tetraprenyl chain at N-7 in the hypoxanthine core were established by extensive spectroscopic analyses. Compounds 1-6 were tested for their inhibitory activity against the phosphodiesterases (PDEs)-4D, 5A, and 9A, and compounds 1 and 6 exhibited moderate inhibitory activity against PDE4D with IC50 values of 8.5 and 20.3 µM, respectively.


Subject(s)
Alkaloids/pharmacology , Anthozoa/chemistry , Phosphodiesterase Inhibitors/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Alkaloids/administration & dosage , Alkaloids/isolation & purification , Animals , China , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 5/drug effects , Inhibitory Concentration 50 , Oceans and Seas , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/isolation & purification , Spectrum Analysis
14.
Respir Res ; 14: 127, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24237854

ABSTRACT

BACKGROUND: Virus-induced exacerbations of Chronic Obstructive Pulmonary Disease (COPD) are a significant health burden and occur even in those receiving the best current therapies. Rhinovirus (RV) infections are responsible for half of all COPD exacerbations. The mechanism by which exacerbations occur remains undefined, however it is likely to be due to virus-induced inflammation. Given that phophodiesterase 4 (PDE4) inhibitors have an anti-inflammatory effect in patients with COPD they present a potential therapy prior to, and during, these exacerbations. METHODS: In the present study we investigated whether the PDE4 inhibitor piclamilast (10(-6) M) could alter RV or viral mimetic (5 µg/mL of imiquimod or poly I:C) induced inflammation and RV replication in primary human airway smooth muscle cells (ASMC) and bronchial epithelial cells (HBEC). The mediators IL-6, IL-8, prostaglandin E2 and cAMP production were assayed by ELISA and RV replication was assayed by viral titration. RESULTS: We found that in ASMCs the TLR3 agonist poly I:C induced IL-8 release was reduced while induced IL-6 release by the TLR7/8 agonist imiquimod was further increased by the presence of piclamilast. However, in RV infected ASMCs, virus replication and induced mediator release were unaltered by piclamilast, as was also found in HBECs. The novel findings of this study reveal that although PDE inhibitors may not influence RV-induced cytokine production in ASMCs and replication in either ASMCs or HBECs, they have the capacity to be anti-inflammatory during TLR activation by modulating the induction of these chemotactic cytokines. CONCLUSION: By extrapolating our in vitro findings to exacerbations of COPD in vivo this suggests that PDE4 inhibitors may have beneficial anti-inflammatory properties when patients are infected with bacteria or viruses other than RV.


Subject(s)
Bronchi/virology , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Cytokines/metabolism , Myocytes, Smooth Muscle/virology , Phosphodiesterase 4 Inhibitors/pharmacology , Pulmonary Disease, Chronic Obstructive/metabolism , Rhinovirus , Toll-Like Receptors/metabolism , Adolescent , Adult , Aged , Aminoquinolines/pharmacology , Benzamides/pharmacology , Bronchi/metabolism , Bronchi/pathology , Cells, Cultured , Comorbidity , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Humans , Imiquimod , In Vitro Techniques , Male , Middle Aged , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Picornaviridae Infections/epidemiology , Picornaviridae Infections/metabolism , Poly I-C/pharmacology , Pulmonary Disease, Chronic Obstructive/epidemiology , Pulmonary Disease, Chronic Obstructive/virology , Pyridines/pharmacology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/drug effects , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/drug effects , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/drug effects , Toll-Like Receptors/drug effects , Young Adult
15.
J Nat Prod ; 76(3): 382-7, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23301853

ABSTRACT

Three bioactive compounds were isolated from an organic extract of an ascomycete fungus of the order Chaetothyriales (MSX 47445) using bioactivity-directed fractionation as part of a search for anticancer leads from filamentous fungi. Of these, two were benzoquinones [betulinan A (1) and betulinan C (3)], and the third was a terphenyl compound, BTH-II0204-207:A (2). The structures were elucidated using a set of spectroscopic and spectrometric techniques; the structure of the new compound (3) was confirmed via single-crystal X-ray diffraction. Compounds 1-3 were evaluated for cytotoxicity against a human cancer cell panel, for antimicrobial activity against Staphylococcus aureus and Candida albicans, and for phosphodiesterase (PDE4B2) inhibitory activities. The putative binding mode of 1-3 with PDE4B2 was examined using a validated docking protocol, and the binding and enzyme inhibitory activities were correlated.


Subject(s)
Ascomycota/chemistry , Benzoquinones/isolation & purification , Benzoquinones/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Phosphodiesterase 4 Inhibitors/isolation & purification , Phosphodiesterase 4 Inhibitors/pharmacology , Staphylococcus aureus/drug effects , Terphenyl Compounds/isolation & purification , Terphenyl Compounds/pharmacology , Ascomycota/classification , Benzoquinones/chemistry , Candida albicans/drug effects , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Drug Screening Assays, Antitumor , Female , Humans , Microbial Sensitivity Tests , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Phosphodiesterase 4 Inhibitors/chemistry , Terphenyl Compounds/chemistry
16.
J Physiol ; 590(2): 309-22, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22083598

ABSTRACT

We tested the hypothesis that inhibition of phosphodiesterase 4 (PDE4) with rolipram to increase vascular endothelial cAMP and stabilize the endothelial barrier would attenuate the action of endogenous atrial natriuretic peptide (ANP) to increase vascular permeability to the plasma protein albumin after an acute plasma volume expansion. After rolipram pretreatment (8 mg (kg body wt)(-1), intraperitoneal, 30 min) more than 95% of the peak increase in plasma volume after volume expansion (4.5% bovine serum albumin, 114 µl (g body wt)(-1) h(-1), 15 min) remained in the vascular space 75 min after the end of infusion, whereas only 67% of the fluid was retained in volume-expanded animals with no rolipram pretreatment. Rolipram significantly decreased 30 min fluorescently labelled albumin clearance (µl (g dry wt)(-1)) relative to untreated volume-expanded controls in skin (e.g. back, 10.4 ± 1.6 vs. 19.5 ± 3.6, P = 0.04), muscle (e.g. hamstring, 15.0 ± 1.9 vs. 20.8 ± 1.4, P = 0.04) and in colon, caecum, and rectum (average reduction close to 50%). The mass of muscle and skin tissue accounted for 70% of volume-expansion-dependent albumin shifts from plasma to interstitium. The results are consistent with observations that the PDE4 inhibitor rolipram attenuates ANP-induced increases in vascular permeability after infusion of exogenous ANP and observations of elevated central venous pressure after a similar volume expansion in mice with selective deletion of the endothelial ANP receptor. These observations may form the basis for new strategies to retain intravenous fluid containing macromolecules.


Subject(s)
Capillary Permeability/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Endothelium, Vascular/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Plasma Volume/drug effects , Rolipram/pharmacology , Animals , Atrial Natriuretic Factor/metabolism , Atrial Natriuretic Factor/pharmacology , Capillary Permeability/physiology , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Central Venous Pressure/drug effects , Central Venous Pressure/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Myocardium/metabolism , Serum Albumin/metabolism
17.
Am J Physiol Renal Physiol ; 303(9): F1307-14, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22933300

ABSTRACT

The thick ascending limb of the loop of Henle (THAL) reabsorbs ∼30% of the filtered NaCl in a process mediated by the apical Na-K-2Cl cotransporter NKCC2. Stimulation of ß-adrenergic receptors in the THAL enhances NaCl reabsorption and increases intracellular cAMP. We found that intracellular cAMP stimulates NKCC2 trafficking to the apical membrane via protein kinase A (PKA). Several cAMP-specific phosphodiesterases (PDE) have been identified in rat THALs, and PDE4 decreases cAMP generated by ß-adrenergic stimulation in other cells. However, it is not known whether ß-adrenergic receptors activation stimulates NKCC2 trafficking. Thus we hypothesized that ß-adrenergic receptor stimulation enhances THAL apical membrane NKCC2 expression via the PKA pathway and PDE4 blunts this effect. THAL suspensions were obtained from Sprague-Dawley rats, and surface NKCC2 expression was measured by surface biotinylation and Western blot. Incubation of THALs with the ß-adrenergic receptor agonist isoproterenol at 0.5 and 1.0 µM increased surface NKCC2 by 17 ± 1 and 29 ± 5% respectively (P < 0.05). Preventing cAMP degradation with 3-isobutyl-methylxanthine (IBMX; a nonselective phosphodiesterase inhibitor) enhanced isoproterenol-stimulated surface NKCC2 expression to 51 ± 7% (P < 0.05 vs. isoproterenol). The ß-adrenergic receptor antagonist propranolol or the PKA inhibitor H-89 completely blocked isoproterenol + IBMX-induced increase on surface NKCC2, while propranolol or H-89 alone had no effect. Selective inhibition of PDE4 with rolipram (20 µM) potentiated the effect of isoproterenol on surface NKCC2 and increased cAMP levels. We concluded that ß-adrenergic receptor stimulation enhances surface NKCC2 expression in the THALs via PKA and PDE4 blunts this effect.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/pharmacology , Loop of Henle/metabolism , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/physiology , Sodium-Potassium-Chloride Symporters/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Enzyme Inhibitors/pharmacology , Isoproterenol/pharmacology , Isoquinolines/pharmacology , Loop of Henle/drug effects , Male , Models, Animal , Phosphodiesterase Inhibitors/pharmacology , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley , Sodium Chloride/metabolism , Sodium-Potassium-Chloride Symporters/drug effects , Solute Carrier Family 12, Member 1 , Sulfonamides/pharmacology
18.
J Pharmacol Exp Ther ; 337(2): 433-43, 2011 May.
Article in English | MEDLINE | ID: mdl-21266552

ABSTRACT

S-Adenosylmethionine (SAM) treatment has anti-inflammatory, cytoprotective effects against endotoxin-induced organ injury. An important component of the anti-inflammatory action of SAM involves down-regulation of the lipopolysaccharide (LPS)-induced transcriptional induction of tumor necrosis factor-α (TNF) expression by monocytes/macrophages. We examined the effect of SAM on expression and activity of LPS-induced up-regulation of phosphodiesterase 4 (PDE4), which regulates cellular cAMP levels and TNF expression. LPS treatment of RAW 264.7, a mouse macrophage cell line, led to the induction of Pde4b2 mRNA expression with no effect on Pde4a or Pde4d. SAM pretreatment led to a significant decrease in LPS-induced up-regulation of Pde4b2 expression in both RAW 264.7 cells and primary human CD14(+) monocytes. Of note, the decreased Pde4b2 mRNA expression correlated with the SAM-dependent increase in the transcriptionally repressive histone H3 lysine 9 trimethylation on the Pde4b2 intronic promoter region. The SAM-mediated decrease in LPS-inducible Pde4b2 up-regulation resulted in an increase in cellular cAMP levels and activation of cAMP-dependent protein kinase A (PKA), which plays an inhibitory role in LPS-induced TNF production. In addition, SAM did not affect LPS-inducible inhibitor of nuclear factor-κB degradation or nuclear factor-κB (NF-κB)-p65 translocation into the nucleus but rather inhibited NF-κB transcriptional activity. These results demonstrate for the first time that inhibition of LPS-induced PDE4B2 up-regulation and increased cAMP-dependent PKA activation are significant mechanisms contributing to the anti-TNF effect of SAM. Moreover, these data also suggest that SAM may be used as an effective PDE4B inhibitor in the treatment of chronic inflammatory disorders in which TNF expression plays a significant pathogenic role.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Phosphodiesterase Inhibitors/pharmacology , S-Adenosylmethionine/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Blotting, Western , Cell Nucleus/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Humans , Lipopolysaccharide Receptors/metabolism , Luciferases/metabolism , Macrophages/drug effects , Macrophages/enzymology , Mice , Monocytes/drug effects , Monocytes/enzymology , Monocytes/metabolism , NF-kappa B/metabolism , Plasmids/genetics , RNA/biosynthesis , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Necrosis Factor-alpha/antagonists & inhibitors , beta-Galactosidase/metabolism
19.
Muscle Nerve ; 44(3): 371-81, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21996797

ABSTRACT

Phosphodiesterase (PDE) inhibition reduces skeletal muscle atrophy, but the underlying molecular mechanism remains unclear. We used microdialysis to investigate the effects of different PDE inhibitors on interstitial tyrosine concentration as well as proteolytic activity and atrogenes expression in isolated rat muscle. Rolipram, a PDE-4-selective inhibitor, reduced the interstitial tyrosine concentration and rates of muscle protein degradation. The rolipram-induced muscle cAMP increase was accompanied by a decrease in ubiquitin-proteasome system (UPS) activity and atrogin-1 mRNA, a ubiquitin-ligase involved in muscle atrophy. This effect was not associated with Akt phosphorylation but was partially blocked by a protein kinase A inhibitor. Fasting increased atrogin-1, MuRF-1 and LC3b expression, and these effects were markedly suppressed by rolipram. Our data suggest that activation of cAMP signaling by PDE-4 blockade leads to inhibition of UPS activity and atrogenes expression independently of Akt. These findings are important for identifying novel approaches to attenuate muscle atrophy.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Gene Expression/drug effects , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Phosphodiesterase 4 Inhibitors/pharmacology , Proteolysis/drug effects , Rolipram/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Gene Expression/physiology , Male , Microtubule-Associated Proteins/metabolism , Models, Animal , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscular Atrophy/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins , Tyrosine/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
20.
Bioorg Med Chem Lett ; 21(21): 6573-6, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21920740

ABSTRACT

A number of novel 1-(3-arylprop-2-ynyl) substituted 1,2-dihydroquinoline derivatives related to nimesulide and their 2-oxo analogues have been designed as potential inhibitors of PDE4. All these compounds were synthesized by using Sonogashira coupling as a key step. In vitro PDE4B inhibitory properties and molecular modeling studies of some of the compounds synthesized are presented.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Quinolines/pharmacology , Sulfonamides/chemistry , Catalytic Domain , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Models, Molecular , Quinolines/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL