Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1867(3): 166019, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33227426

ABSTRACT

Nephritis remains the most common severe manifestation of systemic lupus erythematosus in which auto-antibodies mediate chronic inflammation and kidney damage. cAMP-phosphodiesterases regulate sodium excretion and inflammation in various tissues. How cAMP elevation can reduce systemic inflammation and suppress kidney inflammation and damage remains elusive. PDE4 signaling and cAMP metabolism were investigated along immune complex depositions in target tissues and kidney damage (histology). SLE disease progression is associated with changes in kidney PDE4 activity and expression. Moreover, lupus prone mice exhibit low kidney cAMP level which is associated to induction and relocation of nuclear and cytoskeleton PDE4 isoforms. Auto-antibodies-induced kidney damage was attested by mesangial proliferation and cellular infiltration. Interestingly, we reported that NCS 613 treatment decreases systemic auto-antibody secretion and their corresponding immune complex deposition in target tissues. Furthermore, NCS 613 is able to increase cAMP levels in the kidney; hence this compound rescues kidney PDE4 alterations in treated mice. NCS 613 overcomes disease progression in lupus prone mice by improving wellbeing and decreasing inflammation in treated mice. The PDE4 inhibitor, NCS 613, is a new anti-inflammatory compound that is believed to be a leading drug candidate for the treatment of inflammatory diseases such as lupus nephritis.


Subject(s)
Adenine/analogs & derivatives , Anti-Inflammatory Agents/therapeutic use , Kidney/drug effects , Lupus Nephritis/drug therapy , Phosphodiesterase 4 Inhibitors/therapeutic use , Adenine/pharmacology , Adenine/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Antigen-Antibody Complex/analysis , Antigen-Antibody Complex/immunology , Cyclic AMP/analysis , Cyclic AMP/immunology , Female , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Kidney/immunology , Kidney/pathology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Mice, Inbred MRL lpr , Phosphodiesterase 4 Inhibitors/pharmacology
2.
Front Pharmacol ; 11: 1266, 2020.
Article in English | MEDLINE | ID: mdl-32973507

ABSTRACT

Chronic inflammation is a deleterious process occurring in several pulmonary diseases; it is a driving force promoting tumorigenesis. By regulating local cyclic nucleotide concentration, cyclic nucleotide phosphodiesterases (PDE) govern important biological processes, including inflammation and proliferation. The aim of this study was to investigate the anti-inflammatory and anti-proliferative effects of NCS 613, a specific PDE4 inhibitor, on TNFα-treated human lung adenocarcinoma cell line (A549) and on human lung adenocarcinoma explants. PDE4 isoforms and inflammatory pathways mediated by p38 MAPK, ERK1/2, and IκBα were analyzed by Western blot and immunostainings. Proliferation were performed using [3H]-thymidine incorporation under different experimental conditions. TNFα-stimulation increased p38 MAPK phosphorylation and NF-κB translocation into the nucleus, which was abolished by NCS 613 treatment. Concomitantly, NCS 613 restores IκBα detection level in human adenocarcinoma. An IC50 value of 8.5 µM was determined for NCS 613 on anti-proliferative properties while ERK1/2 signaling was down-regulated in A549 cells and lung adenocarcinoma explants. These findings shed light on PDE4 signaling as a key regulator of chronic inflammation and cancer epithelial cell proliferation. It suggests that PDE4 inhibition by NCS 613 represent potential and interesting strategy for therapeutic intervention in tackling chronic inflammation and cell proliferation.

3.
PLoS One ; 10(12): e0145291, 2015.
Article in English | MEDLINE | ID: mdl-26694325

ABSTRACT

The vasculoprotective properties of delphinidin are driven mainly by its action on endothelial cells. Moreover, delphinidin displays anti-angiogenic properties in both in vitro and in vivo angiogenesis models and thereby might prevent the development of tumors associated with excessive vascularization. This study was aimed to test the effect of delphinidin on melanoma-induced tumor growth with emphasis on its molecular mechanism on endothelial cells. Delphinidin treatment significantly decreased in vivo tumor growth induced by B16-F10 melanoma cell xenograft in mice. In vitro, delphinidin was not able to inhibit VEGFR2-mediated B16-F10 melanoma cell proliferation but it specifically reduced basal and VEGFR2-mediated endothelial cell proliferation. The anti-proliferative effect of delphinidin was reversed either by the MEK1/2 MAP kinase inhibitor, U-0126, or the PI3K inhibitor, LY-294002. VEGF-induced proliferation was reduced either by U-0126 or LY-294002. Under these conditions, delphinidin failed to decrease further endothelial cell proliferation. Delphinidin prevented VEGF-induced phosphorylation of ERK1/2 and p38 MAPK and decreased the expression of the transcription factors, CREB and ATF1. Finally, delphinidin was more potent in inhibiting in vitro cyclic nucleotide phosphodiesterases (PDEs), PDE1 and PDE2, compared to PDE3-PDE5. Altogether delphinidin reduced tumor growth of melanoma cell in vivo by acting specifically on endothelial cell proliferation. The mechanism implies an association between inhibition of VEGF-induced proliferation via VEGFR2 signalling, MAPK, PI3K and at transcription level on CREB/ATF1 factors, and the inhibition of PDE2. In conjunction with our previous studies, we demonstrate that delphinidin is a promising compound to prevent pathologies associated with generation of vascular network in tumorigenesis.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Anthocyanins/administration & dosage , Endothelial Cells/cytology , Melanoma/drug therapy , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Anthocyanins/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Male , Melanoma/metabolism , Melanoma/pathology , Mice , Phosphorylation/drug effects , Xenograft Model Antitumor Assays
4.
Thromb Haemost ; 113(2): 319-28, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25230992

ABSTRACT

Vascular endothelial growth factor (VEGF) plays a major role in angiogenesis by stimulating endothelial cells. Increase in cyclic AMP (cAMP) level inhibits VEGF-induced endothelial cell proliferation and migration. Cyclic nucleotide phosphodiesterases (PDEs), which specifically hydrolyse cyclic nucleotides, are critical in the regulation of this signal transduction. We have previously reported that PDE2 and PDE4 up-regulations in human umbilical vein endothelial cells (HUVECs) are implicated in VEGF-induced angiogenesis and that inhibition of PDE2 and PDE4 activities prevents the development of the in vitro angiogenesis by increasing cAMP level, as well as the in vivo chicken embryo angiogenesis. We have also shown that polyphenols are able to inhibit PDEs. The curcumin having anti-cancer properties, the present study investigated whether PDE2 and PDE4 inhibitors and curcumin could have similar in vivo anti-tumour properties and whether the anti-angiogenic effects of curcumin are mediated by PDEs. Both PDE2/PDE4 inhibitor association and curcumin significantly inhibited in vivo tumour growth in C57BL/6N mice. In vitro, curcumin inhibited basal and VEGF-stimulated HUVEC proliferation and migration and delayed cell cycle progression at G0/G1, similarly to the combination of selective PDE2 and PDE4 inhibitors. cAMP levels in HUVECs were significantly increased by curcumin, similarly to rolipram (PDE4 inhibitor) and BAY-60-550 (PDE2 inhibitor) association, indicating cAMP-PDE inhibitions. Moreover, curcumin was able to inhibit VEGF-induced cAMP-PDE activity without acting on cGMP-PDE activity and to modulate PDE2 and PDE4 expressions in HUVECs. The present results suggest that curcumin exerts its in vitro anti-angiogenic and in vivo anti-tumour properties through combined PDE2 and PDE4 inhibition.


Subject(s)
Angiogenesis Inhibitors/chemistry , Curcumin/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Neoplasms/drug therapy , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase Inhibitors/chemistry , Animals , Cell Cycle , Cell Movement , Cell Proliferation , Cyclic AMP/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/chemistry , Male , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neoplasms/pathology , Rolipram/chemistry , Triazines/chemistry , Vascular Endothelial Growth Factor A/metabolism , Wound Healing
5.
Biochim Biophys Acta ; 1842(7): 916-26, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24631654

ABSTRACT

Production of high titer of antibodies against nuclear components is a hallmark of systemic lupus erythematosus, an autoimmune disease characterized by the progressive chronic inflammation of multiple joints and organs. Organ damage and dysfunction such as renal failure are typical clinical features in lupus. Cell hypermetabolism and hypertrophy can accelerate organ dysfunction. In this study we focus on a specific murine model of lupus, the MRL/lpr strain, and investigated the role of cyclic guanosine monophosphate (cGMP) catabolism in organ remodeling of main target tissues (kidney, spleen and liver) in comparison with age-matched control mice. In MRL/lpr-prone mice, the cGMP-phosphodiesterase (PDE) activities were significantly increased in the kidney (3-fold, P<0.001), spleen (2-fold, P<0.001) and liver (1.6-fold, P<0.05). These raised activity levels were paralleled by both an increased activity of PDE1 in the kidney (associated with nephromegaly) and in the liver, and PDE2 in the spleen of lupus-prone mice. The up-regulation of PDE1 and PDE2 activities were associated with a decrease in intracellular cGMP levels. This underlines an alteration of cGMP-PDE signaling in the kidney, spleen and liver targeting different PDEs according to organs. In good agreement with these findings, a single intravenous administration to MRL/lpr mice of nimodipine (PDE1 inhibitor) but not of EHNA (PDE2 inhibitor) was able to significantly lower peripheral hypercellularity (P=0.0401), a characteristic feature of this strain of lupus-prone mice. Collectively, our findings are important for generating personalized strategies to prevent certain forms of the lupus disease as well as for understanding the role of PDEs and cGMP in the pathophysiology of lupus.


Subject(s)
Cyclic GMP/metabolism , Lupus Erythematosus, Systemic/metabolism , Animals , Female , Lupus Erythematosus, Systemic/genetics , Mice , Mice, Inbred CBA , Mice, Inbred MRL lpr , Up-Regulation
6.
PLoS One ; 7(1): e28899, 2012.
Article in English | MEDLINE | ID: mdl-22247763

ABSTRACT

Systemic lupus erythematosus is a polymorphic and multigenic inflammatory autoimmune disease. Cyclic AMP (cAMP) modulates inflammation and the inhibition of cyclic nucleotide phosphodiesterase type 4 (PDE4), which specifically hydrolyzes cAMP, inhibits TNFα secretion. This study was aimed at investigating the evolution of PDE activity and expression levels during the course of the disease in MRL/lpr lupus-prone mice, and to evaluate in these mice the biological and clinical effects of treatments with pentoxifylline, denbufylline and NCS 613 PDE inhibitors. This study reveals that compared to CBA/J control mice, kidney PDE4 activity of MRL/lpr mice increases with the disease progression. Furthermore, it showed that the most potent and selective PDE4 inhibitor NCS 613 is also the most effective molecule in decreasing proteinuria and increasing survival rate of MRL/lpr mice. NCS 613 is a potent inhibitor, which is more selective for the PDE4C subtype (IC50= 1.4 nM) than the other subtypes (PDE4A, IC50= 44 nM; PDE4B, IC50= 48 nM; and PDE4D, IC50= 14 nM). Interestingly, its affinity for the High Affinity Rolipram Binding Site is relatively low (K(i) = 148 nM) in comparison to rolipram (K(i) = 3 nM). Finally, as also observed using MRL/lpr peripheral blood lymphocytes (PBLs), NCS 613 inhibits basal and LPS-induced TNFα secretion from PBLs of lupus patients, suggesting a therapeutic potential of NCS 613 in systemic lupus. This study reveals that PDE4 represent a potential therapeutic target in lupus disease.


Subject(s)
Adenine/analogs & derivatives , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/mortality , Phosphodiesterase 4 Inhibitors/therapeutic use , Proteinuria/drug therapy , Adenine/therapeutic use , Animals , Cyclic AMP/metabolism , Disease Progression , Female , Humans , Isoenzymes , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Lipopolysaccharides/pharmacology , Lupus Erythematosus, Systemic/enzymology , Mice , Mice, Inbred CBA , Mice, Inbred MRL lpr , Pentoxifylline/therapeutic use , Proteinuria/etiology , Proteinuria/mortality , Survival Rate , Tumor Necrosis Factor-alpha/metabolism , Xanthines/therapeutic use
7.
Br J Pharmacol ; 165(5): 1288-305, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22014080

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDEs) that specifically inactivate the intracellular messengers cAMP and cGMP in a compartmentalized manner represent an important enzyme class constituted by 11 gene-related families of isozymes (PDE1 to PDE11). Downstream receptors, PDEs play a major role in controlling the signalosome at various levels of phosphorylations and protein/protein interactions. Due to the multiplicity of isozymes, their various intracellular regulations and their different cellular and subcellular distributions, PDEs represent interesting targets in intracellular pathways. Therefore, the investigation of PDE isozyme alterations related to various pathologies and the design of specific PDE inhibitors might lead to the development of new specific therapeutic strategies in numerous pathologies. This manuscript (i) overviews the different PDEs including their endogenous regulations and their specific inhibitors; (ii) analyses the intracellular implications of PDEs in regulating signalling cascades in pathogenesis, exemplified by two diseases affecting cell cycle and proliferation; and (iii) discusses perspectives for future therapeutic developments.


Subject(s)
Nucleotides, Cyclic/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Signal Transduction/drug effects , Animals , Humans , Isoenzymes , Phosphodiesterase Inhibitors/therapeutic use
8.
Mol Nutr Food Res ; 55(11): 1677-89, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22045655

ABSTRACT

SCOPE: Curcumin inhibits proliferation of many cancer cells. Cyclic nucleotide phosphodiesterases (PDEs), by hydrolyzing intracellular cyclic adenosine-3',5'-monophosphate (cAMP) and/or cyclic guanosine-3',5'-monophosphate (cGMP), play a pivotal role in signalling pathways involved in cell proliferation. Therefore, this study investigated PDE1-5 participations in the anti-proliferative properties of curcumin in B16F10 murine melanoma cells. METHODS AND RESULTS: We report that curcumin inhibits PDE1-5 activities (IC(50) ≅10(-5) M), indicating that curcumin acts as a non-selective PDE inhibitor. In melanoma cells, PDE4 and PDE1 represent the major cAMP-PDEs and cGMP-PDEs activities, respectively. Curcumin treatment decreased PDE1 and PDE4 activities and dose dependently increased intracellular cGMP levels, whereas cAMP levels were unchanged. Curcumin inhibited cell proliferation and cell cycle progression by accumulating cells in the S- and G2/M-phases with enhanced expressions of cyclin-dependent kinase inhibitors. In contrast, expressions of PDE1A, cyclin A and the epigenetic integrator ubiquitin-like containing PHD and Ring Finger domains 1 (UHRF1) and DNA methyltransferase 1 (DNMT1) were decreased by curcumin. Interestingly, PDE1A overexpression increased UHRF1 and DNMT1 expressions and rescued the B16F10 cells from curcumin anti-proliferative effects. Nimodipine, a PDE1 inhibitor, mimicked the curcumin effects. CONCLUSION: Curcumin exerts its anti-cancer property by targeting PDE1 that inhibits melanoma cell proliferation via UHRF1, DNMT1, cyclin A, p21 and p27 regulations. This suggests that natural PDE1 inhibitors present in food might be effective in preventing cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Curcumin/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Melanoma/drug therapy , Nuclear Proteins/metabolism , Phosphodiesterase Inhibitors/pharmacology , Animals , CCAAT-Enhancer-Binding Proteins , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Melanoma/metabolism , Mice , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Ubiquitin-Protein Ligases
9.
Cell Signal ; 23(1): 152-60, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20807569

ABSTRACT

Thymoquinone (TQ), the active principle of Nigella sativa black seeds, has anti-proliferative properties on numerous cancer cell types. Others and we have previously reported that TQ acts as agent that triggers cell cycle arrest and apoptosis through either a p53- or p73-dependent pathway. However, the immediate targets recruited upon TQ-induced cytotoxicity have not yet been clearly identified. We therefore asked whether cyclic nucleotide phosphodiesterases (PDEs) could be involved in TQ-triggered pro-apoptotic reactivity; PDEs are regulators of intracellular levels of cyclic nucleotides and therefore can modulate cAMP and cGMP-dependent cell death pathways. Our results showed that TQ specifically repressed PDE1A expression in the acute lymphoblastic leukemia Jurkat cell line. This effect is concomitant with the previously described sequential deregulation of the expression of the tumor suppressor protein p73 and the epigenetic integrator UHRF1 (Ubiquitin-like, PHD Ring Finger 1). Interestingly, RNA-interference knock-down of PDE1A expression as well as decreased PDE1A expression induced growth inhibition of Jurkat cells, cell cycle arrest and apoptosis through an activation of p73 and a repression of UHRF1. Conversely, PDE1A re-expression counteracted the cellular pro-apoptotic effects of TQ in association with a p73 repression and UHRF1 re-expression. Altogether, our results show that TQ induced an initial down-regulation of PDE1A with a subsequent down-regulation of UHRF1 via a p73-dependent mechanism. This study further proposes that PDE1A might be involved in the epigenetic code inheritance by regulating, via p73, the epigenetic integrator UHRF1. Our findings also suggest that a forced inhibition of PDE1A expression might be a new therapeutic strategy for the management of acute lymphoblastic leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Benzoquinones/pharmacology , CCAAT-Enhancer-Binding Proteins/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Tumor Suppressor Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Down-Regulation , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Jurkat Cells , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , RNA Interference , Tumor Protein p73 , Ubiquitin-Protein Ligases
10.
PLoS One ; 5(12): e14227, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21151982

ABSTRACT

Left ventricular hypertrophy leads to heart failure and represents a high risk leading to premature death. Cyclic nucleotides (cAMP and cGMP) play a major role in heart contractility and cyclic nucleotide phosphodiesterases (PDEs) are involved in different stages of advanced cardiac diseases. We have investigated their contributions in the very initial stages of left ventricular hypertrophy development. Wistar male rats were treated over two weeks by chronic infusion of angiotensin II using osmotic mini-pumps. Left cardiac ventricles were used as total homogenates for analysis. PDE1 to PDE5 specific activities and protein and mRNA expressions were explored.Rats developed arterial hypertension associated with a slight cardiac hypertrophy (+24%). cAMP-PDE4 activity was specifically increased while cGMP-PDE activities were broadly increased (+130% for PDE1; +76% for PDE2; +113% for PDE5) and associated with increased expressions for PDE1A, PDE1C and PDE5A. The cGMP-PDE1 activation by Ca(2+)/CaM was reduced. BNP expression was increased by 3.5-fold, while NOX2 expression was reduced by 66% and AMP kinase activation was increased by 64%. In early cardiac hypertrophy induced by angiotensin II, all specific PDE activities in left cardiac ventricles were increased, favoring an increase in cGMP hydrolysis by PDE1, PDE2 and PDE5. Increased cAMP hydrolysis was related to PDE4. We observed the establishment of two cardioprotective mechanisms and we suggest that these mechanisms could lead to increase intracellular cGMP: i) increased expression of BNP could increase "particulate" cGMP pool; ii) increased activation of AMPK, subsequent to increase in PDE4 activity and 5'AMP generation, could elevate "soluble" cGMP pool by enhancing NO bioavailability through NOX2 down-regulation. More studies are needed to support these assumptions. Nevertheless, our results suggest a potential link between PDE4 and AMPK/NOX2 and they point out that cGMP-PDEs, especially PDE1 and PDE2, may be interesting therapeutic targets in preventing cardiac hypertrophy.


Subject(s)
Angiotensin II/metabolism , Cardiomegaly/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Gene Expression Regulation, Enzymologic , Animals , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Heart Ventricles/pathology , Hydrolysis , Isoenzymes/chemistry , Male , Membrane Glycoproteins/metabolism , Models, Biological , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Rats , Rats, Wistar
11.
Curr Pharm Des ; 16(9): 1114-25, 2010.
Article in English | MEDLINE | ID: mdl-20030615

ABSTRACT

Cyclic nucleotide phosphodiesterase (PDE), that is a multigenic enzyme superfamily ubiquitously distributed in mammalians, mainly contributes to intracellular signaling regulation. Its various isozymes specifically control in a spatio-temporal manner intracellular levels of cAMP and cGMP downstream receptor activation and nearby functional proteins. The PDE superfamily is constituted by 11 gene families (PDE1-PDE11), comprising 21 genes represented by more than 100 mRNA products due to alternative splicing. Among them, PDE3, PDE4 and PDE5 were viewed as therapeutic targets and therefore, due to the successful development of Viagra (sildenafil, potent selective PDE5 inhibitor), the knowledge in PDE field burst out with the help of academic/pharmaceutical collaborations. Organic medicinal chemistry, using crystallographic and docking approaches, has focused its search on the catalytic pocket of PDEs, leaving aside the development of variant subtype specific PDE inhibitors and activators. This review firstly describes the various properties of each PDE isozyme, focusing particularly on their regulatory domains, mainly located in the N-terminus. Thereafter, we review the possible peptidic regulations of PDE activity itself, then the PDE anchoring in macromolecular complexes and finally the direct interaction of PDE with some critical intracellular proteins, such as beta-arrestin, immunophilin and proteins containing SH3-domain. Altogether, it appears that a peptidic approach would be helpful to study the intrinsic PDE regulation of each subfamily, and thereafter the PDE peptidic motifs implicated as well as PDE location in signaling cascades. Taking in account the various regulatory PDE domains could lead to design new peptides to conceive variant specific inhibitors as well as activators in a therapeutical goal.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/drug effects , Amino Acid Motifs , Peptides/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Conserved Sequence , Drug Delivery Systems/methods , Gene Expression Regulation, Enzymologic/drug effects , Humans , Isoenzymes/chemistry , Isoenzymes/drug effects , Isoenzymes/metabolism , Models, Structural , Protein Interaction Domains and Motifs/drug effects
12.
Int J Exp Pathol ; 90(6): 605-14, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19758418

ABSTRACT

Liver cirrhosis is associated with increased nitric oxide (NO) production in the vasculature. We have previously demonstrated that aorta from rats with liver cirrhosis have a reduced relaxant response to NO donors that is corrected by DMPPO, a PDE5-specific inhibitor. Vasodilator responses to DMPPO itself were also reduced in rings from cirrhotic rats. These results supported previous suggestions that upregulation of PDE5 in liver cirrhosis might contribute to renal sodium retention, and consequently modulate vascular reactivity in the context of increased NO production (Tahseldar-Roumieh et al. in Am. J. Physiol. Heart Circ. Physiol. 290, H481-H488, 2006). Here, we investigated the possible alteration in activity and expression of cyclic nucleotide phosphodiesterase PDE1-PDE5 in kidney and vascular tissues in rats 4 weeks after bile duct ligation. The kidney of rats with cirrhosis had increased activity of PDE1 and PDE4 but not PDE5, and increased expression of PDE1A. Unexpectedly and interestingly, there was no change in cirrhotic aorta PDE5, but an increase in PDE3 and PDE4 activity associated with increased expression of PDE3A and PDE3B. Cilostamide, a specific PDE3 inhibitor, corrected the decreased response to an NO donor in isolated aorta from cirrhotic rats, suggesting that the difference in response to NO donors was due to differences in PDE3-induced hydrolysis of cGMP or to cGMP-induced inhibition of PDE3, rather than to differences in PDE5 contribution. In conclusion, these changes in PDE isozymes could greatly contribute to NO desensitization and to the regulation of vascular and renal function in liver cirrhosis.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Aorta/drug effects , Aorta/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Liver Cirrhosis/enzymology , Nitric Oxide Donors/pharmacology , Animals , Bile Ducts , Isoenzymes/metabolism , Kidney/enzymology , Kidney Cortex/enzymology , Ligation , Mesenteric Arteries/enzymology , Phosphodiesterase Inhibitors/pharmacology , Quinolones/pharmacology , Rats , Rats, Sprague-Dawley
13.
Eur J Pharmacol ; 620(1-3): 78-83, 2009 Oct 12.
Article in English | MEDLINE | ID: mdl-19686719

ABSTRACT

The inhibitory effect of the flavonoid dioclein was assessed on purified vascular cyclic nucleotide phosphodiesterase isoforms (EC 3.1.4.17, PDE1-5) in comparison with 8-methoxymethyl-isobutylmethylxanthine (8-MM-IBMX) and vinpocetine which are currently used as PDE1 inhibitors. The mechanism underlying the vasorelaxant effect of dioclein was investigated in human saphenous vein. Dioclein inhibited PDE1 more selectively than vinpocetine and 8-MM-IBMX, with IC(50) values of 2.47+/-0.26 and 1.44+/-0.35 microM, respectively in basal- and calmodulin-activated states. Dioclein behaved as a competitive inhibitor for cGMP hydrolysis by PDE1 in basal- and calmodulin-activated states (K(i)=0.62+/-0.14 and 0.55+/-0.07 microM, respectively), indicating this inhibitory effect to be independent of calmodulin interactions. In addition, dioclein induced a concentration-dependent relaxation of human saphenous vein which was independent on the presence of functional endothelium (EC(50) values of 7.3+/-3.1 and 11+/-2.7 microM, respectively with and without endothelium). 8-MM-IBMX relaxed human saphenous vein with an EC(50)=31+/-16 microM, whereas vinpocetine did not cause any vasorelaxation at concentrations up to 100 microM. Rp-8-pCPT-cGMPS, which inhibits cGMP-dependent protein kinase (PKG), blocked the vasodilator effect of dioclein, whereas H-89, which is a cAMP-dependent protein kinase (PKA) inhibitor, had a minor inhibitory effect. Our data show that dioclein is a potent calmodulin-independent selective inhibitor of PDE1 and that inhibition of PDE1 is involved in the PKG-mediated vasorelaxant effect of dioclein in human saphenous vein. Furthermore, dioclein may represent a new archetype to develop more specific PDE1 inhibitors.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Flavanones/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Saphenous Vein/cytology , Saphenous Vein/physiology , Vasodilation/drug effects , Animals , Cattle , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/isolation & purification , Saphenous Vein/drug effects
14.
Methods Mol Biol ; 307: 63-74, 2005.
Article in English | MEDLINE | ID: mdl-15988055

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDEs), which are ubiquitously distributed in mammalian tissues, play a major role in cell signaling by hydrolyzing cyclic adenosine 3',5'-monophosphate and cyclic guanosine 3',5'-monophosphate. Owing to their diversity, which allows specific distribution at the cellular and subcellular level, PDEs can selectively regulate various cellular functions. We present here a convenient and sensitive radioenzymatic assay for characterizing and determining the contribution of the various PDE families in cell and tissue extracts. This assay is based on the knowledge and use of chosen PDE family-specific inhibitors in order to determine the distinct PDE isozyme contribution in the overall cyclic nucleotide hydrolyzing activity. It can be used to characterize total, cytosolic, and membrane-associated PDE activities, as well as PDEs associated with purified subcellular structures. This approach is useful for comparing data of control and treated extracts and is therefore quite valuable for viewing the PDE status in different physiopathological conditions.


Subject(s)
2',3'-Cyclic-Nucleotide Phosphodiesterases/chemistry , 3',5'-Cyclic-GMP Phosphodiesterases/chemistry , Biological Assay/methods , 2',3'-Cyclic-Nucleotide Phosphodiesterases/antagonists & inhibitors , 2',3'-Cyclic-Nucleotide Phosphodiesterases/isolation & purification , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-GMP Phosphodiesterases/isolation & purification , Animals , Humans , Isoenzymes/chemistry , Isoenzymes/isolation & purification , Phosphodiesterase Inhibitors/pharmacology , Sensitivity and Specificity , Tissue Extracts
15.
Thromb Haemost ; 92(3): 634-45, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15351862

ABSTRACT

Endothelial cell proliferation in response to VEGF plays an important role in physiological and pathological angiogenesis. The role of PDE2 and PDE4 in VEGF-induced proliferation in HUVEC was investigated: 1) VEGF increased cAMP-hydrolytic activity by up-regulating the expression of PDE2 and PDE4 isozymes; 2) VEGF increased progression in cell cycle with an increase in p42/p44 MAP kinase, cyclin A and cyclin D1 expressions and with a decrease in p21 waf1/cip1 and p27 kip1 expressions; 3) EHNA (20 micro M), a selective PDE2 inhibitor, RP73401 (10 micro M), a selective PDE4 inhibitor blocked the VEGF-induced increase in p42/p44 MAP kinase expression; 4) RP73401, but not EHNA, blocked the VEGF-induced increase in cyclin A and decrease in p27 kip1 expressions; 5) EHNA, contrary to RP73401, enhanced the VEGF-induced increase of cyclin A and decrease of p27 kip1. 6) EHNA and RP73401 together blocked the VEGF-induced increase in cyclin D1 and decrease in p21 waf1/cip1 expressions; 7) Inhibition of VEGF-upregulated PDE2 and PDE4 reversed the VEGF-induced alterations in cell cycle protein expression, bringing back endothelial cells to a non-proliferating status. Consequently, PDE2 and PDE4 inhibitions were able to inhibit VEGF-induced endothelial cell proliferation by restoring cell cycle key protein expression, and might thus be useful in excessive angiogenesis. Furthermore, the differences between PDE2 and PDE4 effects may suggest compartmentalized effects.


Subject(s)
Cell Cycle Proteins/genetics , Cyclic AMP/metabolism , Endothelium, Vascular/cytology , Phosphoric Diester Hydrolases/metabolism , Vascular Endothelial Growth Factor A/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Carrier Proteins/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 2 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Cyclin A/genetics , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Gene Expression Regulation/drug effects , Humans , Hydrolysis , Intracellular Signaling Peptides and Proteins/genetics , Mitogen-Activated Protein Kinase 1/genetics , Umbilical Veins
16.
Cardiovasc Res ; 59(2): 479-87, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12909331

ABSTRACT

OBJECTIVE: Epidemiologic studies have shown that a diet rich in fruits and vegetables has a beneficial preventive effect for cancer and cardiovascular diseases. The mechanisms of these beneficial effects are not known although there is evidence that polyphenolic compounds in food may be of some benefit. The purpose of this study was to define the effect of delphinidin, a vasoactive polyphenol belonging to the class of anthocyanin, on endothelial cell proliferation and migration as well as on in vivo angiogenesis. METHODS AND RESULTS: Vascular endothelial growth factor-stimulated human umbilical endothelial cell migration and proliferation are potently inhibited by delphinidin. Flow cytometric analysis demonstrates that delphinidin inhibition of proliferation is correlated with the blockade of cell cycle in G(0)/G(1) phase. Western blot analysis shows that delphinidin reverses the vascular endothelial growth factor-induced decrease in expression of cyclin-dependent kinase inhibitor p27(kip1) and the vascular endothelial growth factor-induced increase of cyclin D1 and cyclin A, both being necessary to achieve the G(1)-to-S transition. Furthermore, delphinidin inhibits neovascularisation in vivo in chorioallantoic membrane model. CONCLUSION: Delphinidin overcomes in vitro and in vivo angiogenesis and thus appears promising for the development of an anti-angiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Anthocyanins/pharmacology , Cyclins/metabolism , Endothelium, Vascular/metabolism , Blotting, Western/methods , Cell Cycle/drug effects , Cell Movement/drug effects , Cells, Cultured , Depression, Chemical , Endothelium, Vascular/drug effects , Humans , Signal Transduction , Vascular Endothelial Growth Factor A/pharmacology
17.
Thromb Haemost ; 90(2): 334-43, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12888882

ABSTRACT

Migration and proliferation of endothelial cells in response to VEGF play an important role in angiogenesis associated to pathologies such as atherosclerosis, diabetes and tumor development. Elevation of cAMP in endothelial cells has been shown to inhibit growth factor-induced proliferation. Our hypothesis was that inactivation of cAMP-specific phosphodiesterases (PDEs) would inhibit angiogenesis. The purpose of this study was to evaluate the effect of PDE inhibitors on in vitro and in vivo angiogenesis, using human umbilical vein endothelial cell (HUVEC) and chick chorioallantoic membrane (CAM) models respectively. Here, we report that: 1) PDE2, PDE3, PDE4 and PDE5 are expressed in HUVEC; 2) EHNA (20 microM), PDE2 selective inhibitor, and RP73401 (10 microM), PDE4 selective inhibitor, are able to increase the intracellular cAMP level in HUVEC; 3) EHNA and RP73401 are able to inhibit proliferation, cell cycle progression and migration of HUVEC stimulated by VEGF; 4) these in vitro effects can be mimic by treating HUVEC with the cAMP analogue, 8-Br-cAMP (600 microM); 5) only the association of EHNA and RP73401 inhibits in vivo angiogenesis, indicating that both migration and proliferation must be inhibited. These data strongly suggest that PDE2 and PDE4 represent new potential therapeutic targets in pathological angiogenesis.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Adenine/analogs & derivatives , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Phosphoric Diester Hydrolases/drug effects , Vascular Endothelial Growth Factor A/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenine/pharmacology , Allantois/blood supply , Animals , Benzamides/pharmacology , Cell Cycle/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Chick Embryo , Chorion/blood supply , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2 , Cyclic Nucleotide Phosphodiesterases, Type 4 , Enzyme Inhibitors/pharmacology , Humans , Intracellular Membranes/metabolism , Neovascularization, Physiologic/drug effects , Phosphodiesterase Inhibitors/pharmacology , Pyridines/pharmacology , Umbilical Veins
18.
Arterioscler Thromb Vasc Biol ; 23(6): 1001-7, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12676803

ABSTRACT

OBJECTIVE: Moderate consumption of red wine has a beneficial effect on the cardiovascular system. This study examines whether red wine polyphenolic compounds (RWPCs) affect vascular endothelial growth factor (VEGF) expression, a major angiogenic and proatherosclerotic factor in vascular smooth muscle cells (VSMCs). METHODS AND RESULTS: VEGF mRNA expression was assessed by Northern blot analysis and the release of VEGF by immunoassay in cultured VSMCs. Short-term and long-term exposure of VSMCs to RWPCs inhibited VEGF mRNA expression and release of VEGF in response to platelet-derived growth factor AB (PDGFAB), transforming growth factor-beta1, or thrombin. The PDGFAB-induced expression of VEGF was markedly reduced by SB203580 (inhibitor of p38 mitogen-activated protein kinase [MAPK]), antioxidants, and diphenylene iodonium (inhibitor of flavin-dependent enzymes), slightly reduced by PD98059 (inhibitor of MEK), and not significantly affected by wortmannin (inhibitor of PI-3-kinase) and L-JNKI (inhibitor of JNK). Short-term and long-term treatment of VSMCs with RWPCs markedly reduced PDGFAB-induced production of reactive oxygen species and phosphorylation of p38 MAPK. CONCLUSIONS: These data indicate that RWPCs strongly inhibit growth factor-induced VEGF expression in VSMCs by preventing the redox-sensitive activation of the p38 MAPK pathway. The potential antiangiogenic and antiatherosclerotic properties of RWPCs are likely to contribute to cardiovascular protection by preventing the development of atherosclerotic lesions.


Subject(s)
Flavonoids/pharmacology , Phenols/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Wine , Androstadienes/pharmacology , Animals , Antioxidants/pharmacology , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Onium Compounds/pharmacology , Phosphorylation/drug effects , Platelet-Derived Growth Factor/pharmacology , Polyphenols , Protein Processing, Post-Translational/drug effects , Pyridines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Reactive Oxygen Species/metabolism , Thrombin/pharmacology , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Vascular Endothelial Growth Factor A/genetics , Wortmannin , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL