Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Exp Dermatol ; 28(1): 3-10, 2019 01.
Article in English | MEDLINE | ID: mdl-30332502

ABSTRACT

Atopic dermatitis (AD) is a highly prevalent, chronic inflammatory skin disease that affects children and adults. The pathophysiology of AD is complex and involves skin barrier and immune dysfunction. Many immune cytokine pathways are amplified in AD, including T helper (Th) 2, Th22, Th17 and Th1. Current treatment guidelines recommend topical medications as initial therapy; however, until recently, only two drug classes were available: topical corticosteroids (TCSs) and topical calcineurin inhibitors (TCIs). Several limitations are associated with these agents. TCSs can cause a wide range of adverse effects, including skin atrophy, telangiectasia, rosacea and acne. TCIs can cause burning and stinging, and the prescribing information lists a boxed warning for a theoretical risk of malignancy. Novel medications with new mechanisms of action are necessary to provide better long-term control of AD. Phosphodiesterase 4 (PDE4) regulates cyclic adenosine monophosphate in cells and has been shown to be involved in the pathophysiology of AD, making it an attractive therapeutic target. Several PDE4 inhibitors are in clinical development for use in the treatment of AD, including crisaborole, which recently became the first topical PDE4 inhibitor approved for treatment of mild to moderate AD. This review will further describe the pathophysiology of AD, explain the possible role of PDE4 in AD and review PDE4 inhibitors currently approved or being investigated for use in AD.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/prevention & control , Phosphodiesterase Inhibitors/pharmacology , Acetamides/pharmacology , Boron Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cytokines/metabolism , Dermatitis, Atopic/physiopathology , Drug Approval , Humans , Immune System , Inflammation , Phthalic Acids/pharmacology , Pyridines/pharmacology , Quinazolines/pharmacology , Risk , Skin/pathology , Thalidomide/analogs & derivatives , Thalidomide/pharmacology
2.
Toxicol Appl Pharmacol ; 338: 174-181, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29183759

ABSTRACT

Benzophenone-3 (BP-3), which is extensively used in organic sunscreen, has phototoxic potential in human skin. Phosphodiesterase 4B (PDE4B) has a well-established role in inflammatory responses in immune cells. Currently, it is unknown if PDE4B is associated with BP-3-induced phototoxicity in normal human keratinocytes (NHKs). We found that BP-3 significantly increased PDE4B expression in ultraviolet B (UVB)-irradiated NHKs. Notably, BP-8, a sunscreen agent that shares the 2-hydroxy-4-methoxyphenyl methanone moiety with BP-3, also upregulated PDE4B expression in NHKs. Upon UVB irradiation, BP-3 upregulated the expression of pro-inflammatory factors, such as prostaglandin endoperoxide synthase 2, tumor necrosis factor α, interleukin 8, and S100A7, and downregulated the level of cornified envelope associated proteins, which are important in the development of the epidermal permeability barrier. The additive effects of UVB-activated BP-3 on the expression of both pro-inflammatory mediators and cornified envelope associated proteins were antagonized by treatment with the PDE4 inhibitor rolipram. The BP-3 and UVB co-stimulation-induced PDE4B upregulation and its association with the upregulation of pro-inflammatory mediators and the downregulation of epidermal differentiation markers were confirmed in a reconstituted three dimensional human epidermis model. Therefore, PDE4B has a role in the mechanism of BP-3-induced phototoxicity.


Subject(s)
Benzophenones/toxicity , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Dermatitis, Phototoxic/etiology , Keratinocytes/drug effects , Cyclic AMP/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Dinoprostone/biosynthesis , Humans , Interleukin-8/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Ultraviolet Rays
3.
J Am Soc Nephrol ; 28(2): 532-544, 2017 02.
Article in English | MEDLINE | ID: mdl-27466160

ABSTRACT

Trichorhinophalangeal 1 (Trps1) is a transcription factor essential for epithelial cell morphogenesis during kidney development, but the role of Trps1 in AKI induced by ischemia-reperfusion (I/R) remains unclear. Our study investigated Trps1 expression during kidney repair after acute I/R in rats and explored the molecular mechanisms by which Trps1 promotes renal tubular epithelial cell proliferation. Trps1 expression positively associated with the extent of renal repair after I/R injury. Compared with wild-type rats, rats with knockdown of Trps1 exhibited significantly delayed renal repair in the moderate I/R model, with lower GFR levels and more severe morphologic injury, whereas rats overexpressing Trps1 exhibited significantly accelerated renal repair after severe I/R injury. Additionally, knockdown of Trps1 inhibited and overexpression of Trps1 enhanced the proliferation of renal tubular epithelial cells in rats. Chromatin immunoprecipitation sequencing assays and RT-PCR revealed that Trps1 regulated cAMP-specific 3',5'-cyclic phosphodiesterase 4D (Pde4d) expression. Knockdown of Trps1 decreased the renal protein expression of Pde4d and phosphorylated Akt in rats, and dual luciferase analysis showed that Trps1 directly activated Pde4d transcription. Furthermore, knockdown of Pde4d or treatment with the phosphatidylinositol 3 kinase inhibitor wortmannin significantly inhibited Trps1-induced tubular cell proliferation in vitro Trps1 may promote tubular cell proliferation through the Pde4d/phosphatidylinositol 3 kinase/AKT signaling pathway, suggesting Trps1 as a potential therapeutic target for kidney repair after I/R injury.


Subject(s)
Acute Kidney Injury/enzymology , Acute Kidney Injury/pathology , Cell Proliferation , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , DNA-Binding Proteins/physiology , Kidney Tubules/cytology , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Transcription Factors/physiology , Animals , Male , Rats , Rats, Sprague-Dawley , Repressor Proteins
4.
J Cell Sci ; 127(Pt 5): 1033-42, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24413164

ABSTRACT

Multiple cAMP phosphodiesterase (PDE) isoforms play divergent roles in cardiac homeostasis but the molecular basis for their non-redundant function remains poorly understood. Here, we report a novel role for the PDE4B isoform in ß-adrenergic (ßAR) signaling in the heart. Genetic ablation of PDE4B disrupted ßAR-induced cAMP transients, as measured by FRET sensors, at the sarcolemma but not in the bulk cytosol of cardiomyocytes. This effect was further restricted to a subsarcolemmal compartment because PDE4B regulates ß1AR-, but not ß2AR- or PGE2-induced responses. The spatially restricted function of PDE4B was confirmed by its selective effects on PKA-mediated phosphorylation patterns. PDE4B limited the PKA-mediated phosphorylation of key players in excitation-contraction coupling that reside in the sarcolemmal compartment, including L-type Ca(2+) channels and ryanodine receptors, but not phosphorylation of distal cytosolic proteins. ß1AR- but not ß2AR-ligation induced PKA-dependent activation of PDE4B and interruption of this negative feedback with PKA inhibitors increased sarcolemmal cAMP. Thus, PDE4B mediates a crucial PKA-dependent feedback that controls ß1AR-dependent cAMP signals in a restricted subsarcolemmal domain. Disruption of this feedback augments local cAMP/PKA signals, leading to an increased intracellular Ca(2+) level and contraction rate.


Subject(s)
Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Myocytes, Cardiac/enzymology , Receptors, Adrenergic, beta-1/metabolism , Sarcolemma/enzymology , Adrenergic beta-2 Receptor Antagonists/pharmacology , Animals , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Feedback, Physiological , Imidazoles/pharmacology , Myocardial Contraction , Phosphorylation , Protein Processing, Post-Translational , Rats , Receptors, Adrenergic, beta-2/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Second Messenger Systems
5.
Int J Clin Pharmacol Ther ; 53(10): 828-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26308168

ABSTRACT

OBJECTIVE: Phosphodiesterase (PDE) plays an important role in the pathogenesis of Alzheimer's disease (AD). Ferulic acid (FA) has a therapeutic benefit in the treatment of AD. We investigated whether this therapeutic effect is based on the modulation of the PDE/cyclic adenosine monophosphate (cAMP) pathway. In the present study, we investigated whether FA could abrogate Aß25-35- and lipopolysaccharide-induced cellular damage. MATERIALS AND METHODS: Cell viability, superoxide production, and the levels of inflammatory factors were investigated. We further investigated the intracellular levels of cAMP and Ca2+, both of which are associated with PDE activity. Furthermore, molecular docking was used to identify the binding mode between phosphodiesterase 4B2 (PDE4B2) and FA. RESULTS: Pretreatment with FA significantly maintained cell viability, increased the levels of superoxide dismutase, and inhibited production of TNF-α and IL-1ß induced by Aß25-35. Moreover, pretreatment with FA increased the intracellular levels of cAMP and decreased the intracellular levels of Ca2+. The docking results also showed that FA has the potential to inhibit PDE4B2 activity. CONCLUSIONS: Taken together, our results suggested that one of the therapeutic effects of FA on AD was potentially mediated by modulating the PDE/cAMP pathway.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/toxicity , Coumaric Acids/pharmacology , Lipopolysaccharides/toxicity , Peptide Fragments/toxicity , Phosphodiesterase Inhibitors/pharmacology , Animals , Calcium/metabolism , Cell Survival/drug effects , Coumaric Acids/therapeutic use , Cyclic AMP/analysis , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Interleukin-1beta/antagonists & inhibitors , Molecular Docking Simulation , PC12 Cells , Rats , Superoxide Dismutase/biosynthesis , Tumor Necrosis Factor-alpha/antagonists & inhibitors
6.
J Cell Sci ; 125(Pt 21): 5084-95, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22946044

ABSTRACT

Specificity and versatility in cyclic AMP (cAMP) signalling are governed by the spatial localisation and temporal dynamics of the signal. Phosphodiesterases (PDEs) are important for shaping cAMP signals by hydrolyzing the nucleotide. In pancreatic ß-cells, glucose triggers sub-plasma-membrane cAMP oscillations, which are important for insulin secretion, but the mechanisms underlying the oscillations are poorly understood. Here, we investigated the role of different PDEs in the generation of cAMP oscillations by monitoring the concentration of cAMP in the sub-plasma-membrane space ([cAMP](pm)) with ratiometric evanescent wave microscopy in MIN6 cells or mouse pancreatic ß-cells expressing a fluorescent translocation biosensor. The general PDE inhibitor IBMX increased [cAMP](pm), and whereas oscillations were frequently observed at 50 µM IBMX, 300 µM-1 mM of the inhibitor caused a stable increase in [cAMP](pm). The [cAMP](pm) was nevertheless markedly suppressed by the adenylyl cyclase inhibitor 2',5'-dideoxyadenosine, indicating IBMX-insensitive cAMP degradation. Among IBMX-sensitive PDEs, PDE3 was most important for maintaining a low basal level of [cAMP](pm) in unstimulated cells. After glucose induction of [cAMP](pm) oscillations, inhibitors of PDE1, PDE3 and PDE4 inhibitors the average cAMP level, often without disturbing the [cAMP](pm) rhythmicity. Knockdown of the IBMX-insensitive PDE8B by shRNA in MIN6 cells increased the basal level of [cAMP](pm) and prevented the [cAMP](pm)-lowering effect of 2',5'-dideoxyadenosine after exposure to IBMX. Moreover, PDE8B-knockdown cells showed reduced glucose-induced [cAMP](pm) oscillations and loss of the normal pulsatile pattern of insulin secretion. It is concluded that [cAMP](pm) oscillations in ß-cells are caused by periodic variations in cAMP generation, and that several PDEs, including PDE1, PDE3 and the IBMX-insensitive PDE8B, are required for shaping the sub-membrane cAMP signals and pulsatile insulin release.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/physiology , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/physiology , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Insulin/metabolism , Animals , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Female , Glucose/physiology , Insulin Secretion , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Isoenzymes/physiology , Kinetics , Mice , Mice, Inbred C57BL , Periodicity , Phosphatidylinositol Phosphates/metabolism , Second Messenger Systems , Single-Cell Analysis
7.
Blood ; 120(22): 4432-40, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-22995898

ABSTRACT

Ap3A is a platelet-dense granule component released into the extracellular space during the second wave of platelet aggregation on activation. Here, we identify an uncharacterized enzyme, nucleotide pyrophosphatase/phosphodiesterase-4 (NPP4), as a potent hydrolase of Ap3A capable of stimulating platelet aggregation and secretion. We demonstrate that NPP4 is present on the surface of vascular endothelium, where it hydrolyzes Ap3A into AMP and ADP, and Ap4A into AMP and ATP. Platelet aggregation assays with citrated platelet-rich plasma reveal that the primary and secondary waves of aggregation and dense granule release are strongly induced by nanomolar NPP4 in a concentration-dependent manner in the presence of Ap3A, while Ap3A alone initiates a primary wave of aggregation followed by rapid disaggregation. NPP2 and an active site NPP4 mutant, neither of which appreciably hydrolyzes Ap3A, have no effect on platelet aggregation and secretion. Finally, by using ADP receptor blockade we confirm that NPP4 mediates platelet aggregation via release of ADP from Ap3A and activation of ADP receptors. Collectively, these studies define the biologic and enzymatic basis for NPP4 and Ap3A activity in platelet aggregation in vitro and suggest that NPP4 promotes hemostasis in vivo by augmenting ADP-mediated platelet aggregation at the site of vascular injury.


Subject(s)
Blood Coagulation , Endothelium, Vascular/enzymology , Phosphoric Diester Hydrolases/physiology , Pyrophosphatases/physiology , Adenosine Diphosphate/metabolism , Adult , Animals , Blood Coagulation/drug effects , Blood Coagulation/physiology , Coagulants/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Dinucleoside Phosphates/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/injuries , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique , Humans , Hydrolysis , In Vitro Techniques , Insecta , Phosphoric Diester Hydrolases/metabolism , Phosphoric Diester Hydrolases/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Pyrophosphatases/metabolism , Pyrophosphatases/pharmacology , Tissue Distribution
8.
J Biol Chem ; 287(44): 37552-63, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22952227

ABSTRACT

Transient receptor potential vanilloid 1 (TRPV1) is a nonselective cation channel activated by multiple stimuli and is implicated in a variety of pain disorders. Dynamic sensitization of TRPV1 activity by A-kinase anchoring protein 150 demonstrates a critical role for scaffolding proteins in nociception, yet few studies have investigated scaffolding proteins capable of mediating receptor desensitization. In this study, we identify ß-arrestin-2 as a scaffolding protein that regulates TRPV1 receptor activity. We report ß-arrestin-2 association with TRPV1 in multiple cell models. Moreover, siRNA-mediated knockdown of ß-arrestin-2 in primary cultures resulted in a significant increase in both initial and repeated responses to capsaicin. Electrophysiological analysis further revealed significant deficits in TRPV1 desensitization in primary cultures from ß-arrestin-2 knock-out mice compared with wild type. In addition, we found that ß-arrestin-2 scaffolding of phosphodiesterase PDE4D5 to the plasma membrane was required for TRPV1 desensitization. Importantly, inhibition of PDE4D5 activity reversed ß-arrestin-2 desensitization of TRPV1. Together, these results identify a new endogenous scaffolding mechanism that regulates TRPV1 ligand binding and activation.


Subject(s)
Arrestins/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Sensory Receptor Cells/physiology , TRPV Cation Channels/metabolism , Animals , Arrestins/genetics , Arrestins/metabolism , CHO Cells , Cricetinae , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Gene Knockdown Techniques , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Phosphorylation , Primary Cell Culture , Protein Binding , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , TRPV Cation Channels/physiology , Tissue Culture Techniques , Trigeminal Ganglion/cytology , beta-Arrestin 2 , beta-Arrestins
9.
J Pharmacol Exp Ther ; 347(1): 80-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23887098

ABSTRACT

Anti-inflammatory and antifibrotic effects of the broad spectrum phosphodiesterase (PDE) inhibitor pentoxifylline have suggested an important role for cyclic nucleotides in the pathogenesis of hepatic fibrosis; however, studies examining the role of specific PDEs are lacking. Endotoxemia and Toll-like receptor 4 (TLR4)-mediated inflammatory and profibrotic signaling play a major role in the development of hepatic fibrosis. Because cAMP-specific PDE4 critically regulates lipopolysaccharide (LPS)-TLR4-induced inflammatory cytokine expression, its pathogenic role in bile duct ligation-induced hepatic injury and fibrogenesis in Sprague-Dawley rats was examined. Initiation of cholestatic liver injury and fibrosis was accompanied by a significant induction of PDE4A, B, and D expression and activity. Treatment with the PDE4-specific inhibitor rolipram significantly decreased liver PDE4 activity, hepatic inflammatory and profibrotic cytokine expression, injury, and fibrosis. At the cellular level, in relevance to endotoxemia and inflammatory cytokine production, PDE4B was observed to play a major regulatory role in the LPS-inducible tumor necrosis factor (TNF) production by isolated Kupffer cells. Moreover, PDE4 expression was also involved in the in vitro activation and transdifferentiation of isolated hepatic stellate cells (HSCs). Particularly, PDE4A, B, and D upregulation preceded induction of the HSC activation marker α-smooth muscle actin (α-SMA). In vitro treatment of HSCs with rolipram effectively attenuated α-SMA, collagen expression, and accompanying morphologic changes. Overall, these data strongly suggest that upregulation of PDE4 expression during cholestatic liver injury plays a potential pathogenic role in the development of inflammation, injury, and fibrosis.


Subject(s)
Bile Duct Diseases/prevention & control , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Liver Cirrhosis, Experimental/pathology , Phosphodiesterase 4 Inhibitors/therapeutic use , Rolipram/therapeutic use , Up-Regulation/physiology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/physiology , Animals , Bile Duct Diseases/enzymology , Bile Duct Diseases/pathology , Bile Ducts/metabolism , Bile Ducts/pathology , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Ligation , Liver Cirrhosis, Experimental/chemically induced , Male , Phosphodiesterase 4 Inhibitors/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Rolipram/metabolism , Rolipram/pharmacology
10.
Med Sci (Paris) ; 29(6-7): 617-22, 2013.
Article in French | MEDLINE | ID: mdl-23859516

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDE) represent a superfamily of enzymes specialised in the degradation of cAMP and cGMP. In heart, PDE3 and PDE4 are the two major families involved in the regulation of cAMP levels and the control of inotropism. Both families are encoded by several genes, and the recent analysis of the cardiac phenotype of mice lacking these different genes provided new insights into the way they regulate excitation-contraction coupling (ECC). In particular, these studies emphasize the local character of ECC regulation by PDE, as well as the role of these PDE in maintaining calcium homeostasis and preventing cardiac arrhythmias.


Subject(s)
Arrhythmias, Cardiac/etiology , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Myocardial Contraction/physiology , Animals , Arrhythmias, Cardiac/physiopathology , Humans , Mice , Muscle Cells/physiology
11.
Glia ; 60(12): 1839-59, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22865690

ABSTRACT

Cyclic AMP suppresses immune cell activation and inflammation. The positive feedback loop of proinflammatory cytokine production and immune activation implies that cytokines may not only be regulated by cyclic AMP but also conversely regulate cyclic AMP. This study examined the effects of tumor necrosis factor (TNF)-α and interleukin (IL)-1ß on cyclic AMP-phosphodiesterase (PDE) signaling in microglia in vitro and after spinal cord injury (SCI) or traumatic brain injury (TBI). TNF-α or IL-1ß stimulation produced a profound reduction (>90%) of cyclic AMP within EOC2 microglia from 30 min that then recovered after IL-1ß but remained suppressed with TNF-α through 24 h. Cyclic AMP was also reduced in TNF-α-stimulated primary microglia, albeit to a lesser extent. Accompanying TNF-α-induced cyclic AMP reductions, but not IL-1ß, was increased cyclic AMP-PDE activity. The role of PDE4 activity in cyclic AMP reductions was confirmed by using Rolipram. Examination of pde4 mRNA revealed an immediate, persistent increase in pde4b with TNF-α; IL-1ß increased all pde4 mRNAs. Immunoblotting for PDE4 showed that both cytokines increased PDE4A1, but only TNF-α increased PDE4B2. Immunocytochemistry revealed PDE4B nuclear translocation with TNF-α but not IL-1ß. Acutely after SCI/TBI, where cyclic AMP levels are reduced, PDE4B was localized to activated OX-42(+) microglia; PDE4B was absent in OX-42(+) cells in uninjured spinal cord/cortex or inactive microglia. Immunoblotting showed PDE4B2 up-regulation from 24 h to 1 wk post-SCI, the peak of microglia activation. These studies show that TNF-α and IL-1ß differentially affect cyclic AMP-PDE signaling in microglia. Targeting PDE4B2 may be a putative therapeutic direction for reducing microglia activation in CNS injury and neurodegenerative diseases.


Subject(s)
Cyclic AMP/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Cytokines/physiology , Microglia/enzymology , Signal Transduction/physiology , Spinal Cord Injuries/enzymology , Animals , Cell Line , Cells, Cultured , Central Nervous System Diseases/enzymology , Central Nervous System Diseases/pathology , Inflammation Mediators/physiology , Male , Mice , Microglia/pathology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology
12.
Adv Exp Med Biol ; 758: 287-94, 2012.
Article in English | MEDLINE | ID: mdl-23080174

ABSTRACT

UNLABELLED: The cAMP-protein kinase A (PKA) signaling pathway is involved in regulating the release of transmitters from neurons and other cells. Multiple phosphodiesterase (PDE) isoforms regulate this pathway, however, the pattern of isoform expression and stimulus response across tissues has not been fully characterized.Using fluorescent resonance energy transfer (FRET)-based imaging in primary superior cervical ganglia (SCG) neurons and real-time qPCR, we explored the role of PDE3 and PDE4 isoforms and oxygen tension in the activation of PKA and changes in gene expression. These primary neurons were infected with an adenovirus containing A-Kinase activity reporter (AKAR3) and assayed for responses to PDE inhibitors: rolipram (ROL, 1 µM), milrinone (MIL, 10 µM) and IBMX (100 µM), and adenylyl cyclase activator forskolin (FSK, 50 µM). Different PDE activity patterns were observed in different cells: high PDE4 activity (n = 3), high PDE3 activity (n = 3) and presence of activity of other PDEs (n = 3). Addition of PKA inhibitor H89 (10 µM) completely reversed the response. We further studied the effect of oxygen in the PKA activity induced by PDE inhibition. Both normoxia (20%O(2)/5%CO(2)) and hypoxia (0%O(2)/5%CO(2)) induced a similar increase in the FRET emission ratio (14.5 ± 0.8 and 14.7 ± 0.8, respectively).PDE3a, PDE4b and PDE4d isoforms mRNAs were highly expressed in the whole SCG with no modulation by hypoxia. CONCLUSION: Using a FRET-based PKA activity sensor, we show that primary SCG neurons can be used as a model system to dissect the contribution of different PDE isoforms in regulating cAMP/PKA signaling. The differential patterns of PDE regulation potentially represent subpopulations of ganglion cells with different physiological functions.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Oxygen/physiology , Superior Cervical Ganglion/enzymology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Female , Fluorescence Resonance Energy Transfer , Isoenzymes/genetics , Isoenzymes/physiology , Male , Rats , Rats, Sprague-Dawley
13.
Proc Natl Acad Sci U S A ; 106(51): 21918-23, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-19955404

ABSTRACT

Beta-arrestins, key regulators of receptor signaling, are highly expressed in the central nervous system, but their roles in brain physiology are largely unknown. Here we show that beta-arrestin-2 is critically involved in the formation of associative fear memory and amygdalar synaptic plasticity. In response to fear conditioning, beta-arrestin-2 translocates to amygdalar membrane where it interacts with PDE-4, a cAMP-degrading enzyme, to inhibit PKA activation. Arrb2(-/-) mice exhibit impaired conditioned fear memory and long-term potentiation at the lateral amygdalar synapses. Moreover, expression of the beta-arrestin-2 in the lateral amygdala of Arrb2(-/-) mice, but not its mutant form that is incapable of binding PDE-4, restores basal PKA activity and rescues conditioned fear memory. Taken together, our data demonstrate that the feedback regulation of amygdalar PKA activation by beta-arrestin-2 and PDE-4 complex is critical for the formation of conditioned fear memory.


Subject(s)
Amygdala/enzymology , Arrestins/physiology , Conditioning, Operant , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Fear , Animals , Arrestins/metabolism , Biofeedback, Psychology , Blotting, Western , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Immunoprecipitation , Mice , Mice, Knockout , beta-Arrestin 2 , beta-Arrestins
14.
J Cell Physiol ; 226(8): 1970-80, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21520048

ABSTRACT

Lung fibroblast proliferation and differentiation into myofibroblasts are pathological key events during development of lung fibrosis. Cyclic nucleotide signaling is described as a negative modulator of these cellular processes, and cyclic nucleotide degrading type 4 phosphodiesterases (PDE4) are important regulators of these pathways. In this study, we elucidated expression and the role of individual subtypes of PDE4 in primary normal human lung fibroblast (NHLF) in controlling cytokines-induced proliferation and conversion to myofibroblasts by short-interfering RNAs (siRNAs) induced knockdown. We verified the expression of PDE4A, B, and D, while PDE4C was only minor or even not expressed in NHLF. An efficient liposome-mediated transfection method for mRNA silencing and a knockdown of the expressed PDE4 subtypes was achieved in these cells. This knockdown was further validated by PDE4 protein expression analysis and PDE4 activity measurements. Functionally, the knockdown of PDE4A and PDE4B inhibited proliferation induced by the cytokine combination of bFGF and IL-1ß, whereas knockdown of PDE4D was ineffective. In contrast, TGF-ß induced differentiation into myofibroblasts was affected by knockdown of PDE4B and PDE4D, but not by PDE4A knockdown. In summary, our data allow to assign different PDE4 subtypes to distinct functions of human lung fibroblasts and highlight the predominant role of PDE4B in controlling pathophysiological processes of human lung fibroblasts. This provides a scientific rationale for focused therapeutic targeting of PDE4B to treat respiratory diseases with fibrotic lesions in the lung.


Subject(s)
Cell Proliferation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Cytokines/metabolism , Fibroblasts/enzymology , Lung/enzymology , Myofibroblasts/enzymology , Aminopyridines/pharmacology , Benzamides/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cyclopropanes/pharmacology , Cytokines/physiology , Fibroblast Growth Factor 2/metabolism , Gene Expression Profiling , Humans , Interleukin-1beta/metabolism , Isoenzymes/metabolism , Lung/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Pyridines/pharmacology , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Transfection , Transforming Growth Factor beta/metabolism
15.
Ann Rheum Dis ; 70(6): 1130-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21342916

ABSTRACT

BACKGROUND: The cAMP-metabolising enzyme, phosphodiesterase 4 (PDE4), has been implicated in a number of immune responses, including tumour necrosis factor α (TNFα) production. To date, few data have directly addressed whether synovial cytokine and chemokine production is modified by PDE4. OBJECTIVE: Using specific PDE4 inhibitors, roflumilast plus two novel inhibitors, INH 0061 and INH 0062, the authors studied the effect of PDE4 inhibition on proinflammatory cytokine and chemokine release from primary rheumatoid arthritis (RA) synovial digest suspensions and in a macrophage T cell co-culture assay system. RESULTS: All PDE4 inhibitors dose-dependently reduced the release of TNFα from primary synovial membrane cultures (n=5), half maximal inhibitory concentration (IC(50)) 300-30 nM, p<0.05. Similarly, a significant suppression in the release the proinflammatory chemokines, monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory protein (MIP)-1α, MIP-1ß (IC(50) 300-30 nM) and regulated upon activation normal T-cell expressed and secreted (RANTES) (IC(50) 3 nM) was also observed, p<0.05. While interleukin 1ß was also reduced, it did not achieve an IC(50). These observations were further confirmed in a macrophage T cell co-culture system, demonstrating the importance of PDE4 pathways in regulating cytokine/chemokine release in a cellular interaction implicated in inflammatory synovitis. Subsequent studies using the human monocytic cell line U937 also demonstrated cytokine regulation with PDE4 knockdown utilising a small interfering RNA approach. CONCLUSION: These data provide direct evidence of PDE4-dependent pathways in human RA synovial inflammatory cytokine and chemokine release and may provide a novel approach in treating chronic autoimmune conditions such as RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Cytokines/metabolism , Inflammation Mediators/metabolism , Synovial Membrane/immunology , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/pathology , Cell Communication/immunology , Cells, Cultured , Coculture Techniques , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Cytokines/biosynthesis , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Humans , Phosphodiesterase 4 Inhibitors/pharmacology , RNA, Small Interfering/genetics , Synovial Membrane/drug effects , Synovial Membrane/enzymology , Synovitis/enzymology , Synovitis/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
16.
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
17.
Handb Exp Pharmacol ; (204): 391-414, 2011.
Article in English | MEDLINE | ID: mdl-21695650

ABSTRACT

Phosphodiesterase 4 (PDE4) belongs to a family of enzymes which catalyzes the breakdown of 3, 5'-adenosine cyclic monophosphate (cAMP) and is ubiquitously expressed in inflammatory cells. There is little evidence that inflammatory diseases are caused by increased expression of this isoenzyme, although human inflammatory cell activity can be suppressed by selective PDE4 inhibitors. Consequently, there is intense interest in the development of selective PDE4 inhibitors for the treatment of a range of inflammatory diseases, including asthma, chronic obstructive pulmonary disease (COPD), inflammatory bowel disease, and psoriasis. Recent clinical trials with roflumilast in COPD have confirmed the therapeutic potential of targeting PDE4 and recently roflumilast has been approved for marketing in Europe and the USA, although side effects such as gastrointestinal disturbances, particularly nausea and emesis as well as headache and weight loss, may limit the use of this drug class, at least when administered by the oral route. However, a number of strategies are currently being pursued in attempts to improve clinical efficacy and reduce side effects of PDE4 inhibitors, including delivery via the inhaled route, development of nonemetic PDE4 inhibitors, mixed PDE inhibitors, and/or antisense biologicals targeted toward PDE4.


Subject(s)
Inflammation/drug therapy , Phosphodiesterase 4 Inhibitors/therapeutic use , Animals , Asthma/drug therapy , Asthma/etiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Humans , Phosphodiesterase 4 Inhibitors/adverse effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/etiology
18.
J BUON ; 26(4): 1517-1522, 2021.
Article in English | MEDLINE | ID: mdl-34565013

ABSTRACT

PURPOSE: To explore the role of lncRNA MANCR in regulating in vitro proliferation and apoptosis in esophageal carcinoma cells and in vivo growth of esophageal carcinoma in nude mice. METHODS: MANCR levels in 15 pairs of esophageal carcinomas and non-tumoral tissues were detected by qRT-PCR. In vitro regulations of MANCR on proliferative and apoptotic potentials in TE-1 and EC-109 cells were explored by CCK-8, colony formation assay and flow cytometry. In addition, dual-luciferase reporter assay and rescue experiments were conducted to clarify the potential mechanisms of MANCR on regulating PDE4D. Finally, in vivo role of MANCR in mediating esophageal carcinoma growth was determined in nude mice implanted with EC-109 cells. RESULTS: MANCR was highly expressed in esophageal carcinomas tissues than non-tumoral ones. MANCR promoted proliferative ability and inhibited apoptosis in TE-1 and EC-109 cells. In nude mice with xenografted esophageal carcinoma, knockdown of MANCR markedly slowed down tumor growth. PDE4D was the target gene binding MANCR, which was downregulated in esophageal carcinoma tissues. Its level was negatively regulated by MANCR. Importantly, PDE4D could abolish the role of MANCR in stimulating the malignant progression of esophageal carcinoma. CONCLUSIONS: LncRNA MANCR is upregulated in esophageal carcinoma cases. Through negatively regulating PDE4D level, MANCR stimulates proliferative ability and inhibits apoptosis in esophageal carcinoma, thus driving the malignant progression.


Subject(s)
Carcinoma/pathology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Esophageal Neoplasms/pathology , RNA, Long Noncoding/physiology , Animals , Disease Progression , Male , Mice , Mice, Nude
19.
J Sex Med ; 7(6): 2104-2111, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20487244

ABSTRACT

INTRODUCTION: The cyclic adenosine monophosphate-dependent protein kinase (cAK) is considered a key protein in the control of smooth muscle tone in the cardiovascular system. There is evidence that erectile dysfunction might be linked to systemic vascular disorders and arterial insufficiency, subsequently resulting in structural changes in the penile tissue. The expression and significance of cAK in human cavernous arteries (HCA) have not been evaluated. AIMS: To evaluate the expression of cAK isoforms in HCA and examine the role of cAK in the cyclic adenosine monophosphate (cAMP)- and cyclic guanosine monophosphate (cGMP)-mediated control of penile vascular smooth muscle. METHODS: The expression and distribution of phosphodiesterase type 4 (PDE4) and cAK isoforms in sections of HCA were investigated by means of immunohistochemistry and Western blot analysis. The effects of the cAK inhibitor Rp-8-CPT-cAMPS on the relaxation of isolated preparations of HCA (diameter > 100 µm) induced by rolipram, sildenafil, tadalafil, and vardenafil were studied using the organ bath technique. MAIN OUTCOME MEASURES: Investigate the expression of cAK in relation to α-actin and PDE4 in HCA and evaluate the effects of an inhibition of cAK on the relaxation induced by inhibitors of PDE4 and PDE5 of isolated penile arteries. RESULTS: Immunosignals specific for cAKIα, IIα, and IIß were observed within the wall of HCA. Double stainings revealed colocalization of cAK with α-actin and PDE4. The expression of cAK isoforms was confirmed by Western blot analysis. The reversion of tension induced by inhibitors of PDE4 and PDE5 of isolated penile vascular tissue were attenuated significantly by Rp-8-CPT-cAMPS. CONCLUSIONS: Our results demonstrate the expression of cAK isoforms in the smooth musculature of HCA and its colocalization with PDE4. A significant role for cAK in the regulation mediated by cAMP and cGMP of vascular smooth muscle tone in HCA can also be assumed.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Cyclic Nucleotide Phosphodiesterases, Type 5/physiology , Muscle, Smooth, Vascular/physiology , Penis/blood supply , Vasodilation/physiology , Actins/physiology , Adult , Arteries/physiology , Cyclin-Dependent Kinases/physiology , Humans , In Vitro Techniques , Male , Middle Aged , Protein Isoforms/physiology , Signal Transduction/physiology , Cyclin-Dependent Kinase-Activating Kinase
20.
J Neurosci ; 28(42): 10460-71, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18923023

ABSTRACT

Phosphodiesterase (PDE) is a critical regulator of cAMP/protein kinase A (PKA) signaling in cells. Multiple PDEs with different substrate specificities and subcellular localization are expressed in neurons. Dopamine plays a central role in the regulation of motor and cognitive functions. The effect of dopamine is largely mediated through the cAMP/PKA signaling cascade, and therefore controlled by PDE activity. We used in vitro and in vivo biochemical techniques to dissect the roles of PDE4 and PDE10A in dopaminergic neurotransmission in mouse striatum by monitoring the ability of selective PDE inhibitors to regulate phosphorylation of presynaptic [e.g., tyrosine hydroxylase (TH)] and postsynaptic [e.g., dopamine- and cAMP-regulated phosphoprotein of M(r) 32 kDa (DARPP-32)] PKA substrates. The PDE4 inhibitor, rolipram, induced a large increase in TH Ser40 phosphorylation at dopaminergic terminals that was associated with a commensurate increase in dopamine synthesis and turnover in striatum in vivo. Rolipram induced a small increase in DARPP-32 Thr34 phosphorylation preferentially in striatopallidal neurons by activating adenosine A(2A) receptor signaling in striatum. In contrast, the PDE10A inhibitor, papaverine, had no effect on TH phosphorylation or dopamine turnover, but instead robustly increased DARPP-32 Thr34 and GluR1 Ser845 phosphorylation in striatal neurons. Inhibition of PDE10A by papaverine activated cAMP/PKA signaling in both striatonigral and striatopallidal neurons, resulting in potentiation of dopamine D(1) receptor signaling and inhibition of dopamine D(2) receptor signaling. These biochemical results are supported by immunohistochemical data demonstrating differential localization of PDE10A and PDE4 in striatum. These data underscore the importance of individual brain-enriched cyclic-nucleotide PDE isoforms as therapeutic targets for neuropsychiatric and neurodegenerative disorders affecting dopamine neurotransmission.


Subject(s)
Corpus Striatum/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclic AMP/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Phosphoric Diester Hydrolases/physiology , Animals , Corpus Striatum/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL