Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(2): e0263828, 2022.
Article in English | MEDLINE | ID: mdl-35148334

ABSTRACT

BACKGROUND AND AIMS: Nonalcoholic Steatohepatitis (NASH) is a major cause of end-stage liver diseases such as cirrhosis and hepatocellular carcinoma resulting ultimately in increased liver-related mortality. Fibrosis is the main driver of mortality in NASH. Procollagen C-Proteinase Enhancer-1 (PCPE-1) plays a key role in procollagen maturation and collagen fibril formation. To assess its role in liver fibrosis and NASH progression, knock-out mice were evaluated in a dietary NASH model. METHODS: Global constitutive Pcolce-/- and WT male mice were fed with a Choline Deficient Amino acid defined High Fat Diet (CDA HFD) for 8 weeks. Liver triglycerides, steatosis, inflammation and fibrosis were assessed at histological, biochemical and gene expression levels. In addition, human liver samples from control and NASH patients were used to evaluate the expression of PCPE-1 at both mRNA and protein levels. RESULTS: Pcolce gene deficiency prevented diet-induced liver enlargement but not liver dysfunction. Furthermore, liver triglycerides, steatosis and inflammation were not modified in Pcolce-/- male mice compared to WT under CDA HFD. However, a significant decrease in liver fibrosis was observed in Pcolce-/- mice compared to WT under NASH diet, associated with a decrease in total and insoluble collagen content without any significant modifications in the expression of genes involved in fibrosis and extracellular matrix remodeling. Finally, PCPE-1 protein expression was increased in cirrhotic liver samples from both NASH and Hepatitis C patients. CONCLUSIONS: Pcolce deficiency limits fibrosis but not NASH progression in CDA HFD fed mice.


Subject(s)
Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/pathology , Animals , Diet, High-Fat , Disease Models, Animal , Disease Progression , Female , Gene Knockout Techniques , Humans , Liver/chemistry , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Male , Mice , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Triglycerides/chemistry , Up-Regulation
2.
J Comput Chem ; 43(10): 692-703, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35218219

ABSTRACT

Multi-parameter optimization (MPO) is a major challenge in new chemical entity (NCE) drug discovery. Recently, promising results were reported for deep learning generative models applied to de novo molecular design, but, to our knowledge, until now no report was made of the value of this new technology for addressing MPO in an actual drug discovery project. In this study, we demonstrate the benefit of applying AI technology in a real drug discovery project. We evaluate the potential of a ligand-based de novo design technology using deep learning generative models to accelerate the obtention of lead compounds meeting 11 different biological activity objectives simultaneously. Using the initial dataset of the project, we built QSAR models for all the 11 objectives, with moderate to high performance (precision between 0.67 and 1.0 on an independent test set). Our DL-based AI de novo design algorithm, combined with the QSAR models, generated 150 virtual compounds predicted as active on all objectives. Eleven were synthetized and tested. The AI-designed compounds met 9.5 objectives on average (i.e., 86% success rate) versus 6.4 (i.e., 58% success rate) for the initial molecules measured on all objectives. One of the AI-designed molecules was active on all 11 measured objectives, and two were active on 10 objectives while being in the error margin of the assay for the last one. The AI algorithm designed compounds with functional groups, which, although being rare or absent in the initial dataset, turned out to be highly beneficial for the MPO.


Subject(s)
Drug Design , Drug Discovery , Algorithms , Drug Discovery/methods , Ligands
3.
J Cardiovasc Pharmacol ; 78(5): e703-e713, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34369899

ABSTRACT

ABSTRACT: Maturation of fibrillar collagen is known to play a crucial role in the pathophysiology of myocardial fibrosis. Procollagen C-proteinase enhancer 1 (PCPE1) has a key role in procollagen maturation and collagen fibril formation. The phenotype of both male and female PCPE1 knock-out mice was investigated under basal conditions to explore the potential of PCPE1 as a therapeutic target in heart failure. Global constitutive PCPE1-/- mice were generated. Serum procollagen I C-terminal propeptide, organ histology, and cutaneous wound healing were assessed in both wild type (WT) and PCPE1-/- mice. In addition, the cardiac expression of genes involved in collagen metabolism was investigated and the total and insoluble cardiac collagen contents determined. Cardiac function was evaluated by echocardiography. No differences in survival, clinical chemistry, or organ histology were observed in PCPE1-/- mice compared with WT. Serum procollagen I C-terminal propeptide was lower in PCPE1-/- mice. Cardiac mRNA expression of Bmp1, Col1a1, Col3a1, and Loxl2 was similar, whereas Tgfb and Loxl1 mRNA levels were decreased in PCPE1-/- mice compared with sex-matched WT. No modification of total or insoluble cardiac collagen content was observed between the 2 strains. Ejection fraction was slightly decreased in PCPE1-/- male mice, but not in females. Finally, wound healing was not altered in PCPE1-/- mice. PCPE1 deficiency does not trigger any major liabilities and does not affect cardiac collagen content nor its function under basal conditions. Further studies are required to evaluate its role under stressed conditions and determine its suitability as a therapeutic target for heart failure.


Subject(s)
Collagen/metabolism , Extracellular Matrix Proteins/deficiency , Myocardium/metabolism , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Animals , Bone Morphogenetic Protein 1/genetics , Bone Morphogenetic Protein 1/metabolism , Collagen/genetics , Collagen Type I, alpha 1 Chain/genetics , Collagen Type I, alpha 1 Chain/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Extracellular Matrix Proteins/genetics , Female , Gene Expression Regulation , Genotype , Male , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/blood , Phenotype , Procollagen/blood , Stroke Volume , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Ventricular Function, Left , Wound Healing
4.
Transl Res ; 238: 36-48, 2021 12.
Article in English | MEDLINE | ID: mdl-34332154

ABSTRACT

Little is known about the mechanisms of aging on vascular beds and its relationship with tetra and di-hydrobiopterin (BH4 and BH2) levels. This observational clinical study analyzed the impact of aging on plasma and platelet biopterins, cutaneous blood flow (CBF), and coronary flow reserve (CFR) in healthy adults. The study enrolled healthy adults in 3 age groups: 18-30, 50-59, and 60-70 years (n = 25/group). Biopterins were assessed by LC-MS/MS using newly defined pre-analytical conditions limiting BH4 oxidation and improving long-term stability. CBF was measured by Laser Speckle Contrast Imaging coupled with acetylcholine-iontophoresis and CFR by adenosine stress cardiac magnetic resonance. In healthy adults, aging (60-70 years vs 18-30 years) significantly increased platelet BH2 (+75%, P = 0.033) and BH2 + BH4 (+31%, P = 0.033), and to a lesser extent plasma BH2 (+29%, P = 0.009) without affecting BH4 and BH4/BH2. Simultaneously, CBF was decreased (-23%, P = 0.004) but not CFR, CBF being inversely correlated with platelet BH2 (r = -0.42, P = 0.001) and BH2 + BH4 (r = -0.41, P = 0.002). The proportion of adults with abnormal platelet BH2 increased with age (+28% in 60-70y). These abnormal BH2 levels were significantly associated with reduced CBF and CFR (-16%, P = 0.03 and -26%, P = 0.02). In conclusion, our study showed that age-related peripheral endothelial dysfunction was associated with an increase in circulating BH2 without decreasing BH4, the effect being more marked in platelets, the most relevant blood compartment to assess biopterin bioavailability. Peripheral but not coronary vascular function is progressively impaired with aging in healthy adults. All these findings support biopterins as therapeutic targets to improve vascular function.


Subject(s)
Aging/physiology , Biopterins/analogs & derivatives , Endothelium, Vascular/physiopathology , Adolescent , Adult , Aged , Animals , Biopterins/blood , Blood Platelets/metabolism , Blood Vessels/physiology , Endothelium, Vascular/physiology , Female , Humans , Male , Middle Aged , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Zucker , Young Adult
5.
Thromb Res ; 204: 81-87, 2021 08.
Article in English | MEDLINE | ID: mdl-34153648

ABSTRACT

Enhancement of fibrinolysis constitutes a promising approach to treat thrombotic diseases. Venous thrombosis and thromboembolism risks are associated with increased plasma levels of TAFI (Thrombin Activatable Fibrinolysis Inhibitor) as well as its active form TAFIa. A new TAFIa inhibitor, namely S62798 has been identified. Its ability to enhance fibrinolysis was investigated both in vitro and in vivo in a mouse model of pulmonary thromboembolism, as well as its effect on bleeding. S62798 is a highly selective human, mouse and rat TAFIa inhibitor (IC50 = 11; 270; 178 nmol/L, respectively). It accelerates lysis of a human clot in vitro, evaluated by thromboelastometry (EC50 = 27 nmol/L). In a rat tail bleeding model, no effect of S62798 treatment was observed up to 20 mg/kg. Enhancement of endogenous fibrinolysis by S62798 was investigated in a mouse model of Tissue Factor-induced pulmonary thromboembolism. Intravenous administration of S62798 decreased pulmonary fibrin clots with a minimal effective dose of 0.03 mg/kg. Finally, effect of S62798 in combination with heparin was evaluated. When treatment of heparin was done in a curative setting, no effect was observed whereas a significantly decreased pulmonary fibrin deposition was observed in response to S62798 alone or in combination with heparin. This study demonstrates that S62798 is a potent TAFIa inhibitor with minimal risk of bleeding. In vivo, curative S62798 intravenous treatment, alone or associated with heparin, accelerated clot lysis by potentiating endogenous fibrinolysis and thus decreased pulmonary fibrin clots. S62798 is expected to be a therapeutic option for pulmonary embolism patients on top of anticoagulants.


Subject(s)
Carboxypeptidase B2 , Enzyme Inhibitors/pharmacology , Pulmonary Embolism , Animals , Carboxypeptidase B2/antagonists & inhibitors , Disease Models, Animal , Fibrin Clot Lysis Time , Fibrinolysis , Humans , Mice , Pulmonary Embolism/drug therapy , Rats
6.
J Med Chem ; 64(7): 3897-3910, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33764059

ABSTRACT

Selective and potent inhibitors of activated thrombin activatable fibrinolysis inhibitor (TAFIa) have the potential to increase endogenous and therapeutic fibrinolysis and to behave like profibrinolytic agents without the risk of major hemorrhage, since they do not interfere either with platelet activation or with coagulation during blood hemostasis. Therefore, TAFIa inhibitors could be used in at-risk patients for the treatment, prevention, and secondary prevention of stroke, venous thrombosis, and pulmonary embolisms. In this paper, we describe the design, the structure-activity relationship (SAR), and the synthesis of novel, potent, and selective phosphinanes and azaphosphinanes as TAFIa inhibitors. Several highly active azaphosphinanes display attractive properties suitable for further in vivo efficacy studies in thrombosis models.


Subject(s)
Aza Compounds/pharmacology , Carboxypeptidase B2/antagonists & inhibitors , Cyclic P-Oxides/pharmacology , Fibrinolytic Agents/pharmacology , Phosphinic Acids/pharmacology , Protease Inhibitors/pharmacology , Animals , Aza Compounds/chemical synthesis , Aza Compounds/metabolism , Carboxypeptidase B2/metabolism , Catalytic Domain , Cyclic P-Oxides/chemical synthesis , Cyclic P-Oxides/metabolism , Fibrinolysis/drug effects , Fibrinolytic Agents/chemical synthesis , Fibrinolytic Agents/metabolism , Humans , Male , Molecular Docking Simulation , Molecular Structure , Phosphinic Acids/chemical synthesis , Phosphinic Acids/metabolism , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Protein Binding , Rats, Sprague-Dawley , Structure-Activity Relationship
7.
Pflugers Arch ; 467(9): 1945-63, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25385304

ABSTRACT

Angiotensin-II and oxidative stress are involved in the genesis of aortic aneurysms, a phenomenon exacerbated by endothelial nitric oxide synthase (eNOS) deletion or uncoupling. The purpose of this work was to study the endothelial function in wild-type C57BL/6 (BL) and transgenic mice expressing the h-angiotensinogen and h-renin genes (AR) subjected to either a control, or a high-salt diet plus a treatment with a NO-synthase inhibitor, N-ω-nitro-L-arginine-methyl-ester (L-NAME; BLSL and ARSL). BLSL showed a moderate increase in blood pressure, while ARSL became severely hypertensive. Seventy-five percent of ARSL developed aortic aneurysms, characterized by major histo-morphological changes and associated with an increase in NADP(H) oxidase-2 (NOX2) expression. Contractile responses (KCl, norepinephrine, U-46619) were similar in the four groups of mice, and relaxations were not affected in BLSL and AR. However, in ARSL, endothelium-dependent relaxations (acetylcholine, UK-14304) were significantly reduced, and this dysfunction was similar in aortae without or with aneurysms. The endothelial impairment was unaffected by catalase, superoxide-dismutase mimetic, radical scavengers, cyclooxygenase inhibition, or TP-receptor blockade and could not be attributed to sGC oxidation. Thus, ARSL is a severe hypertension model developing aortic aneurysm. A vascular dysfunction, involving both endothelial (reduced role of NO) and smooth muscle cells, precedes aneurysms formation and, paradoxically, does not appear to involve oxidative stress.


Subject(s)
Aorta/physiopathology , Aortic Aneurysm/physiopathology , Endothelium, Vascular/physiopathology , Hypertension/complications , Oxidative Stress/physiology , Animals , Disease Models, Animal , Female , Humans , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Polymerase Chain Reaction , Vasodilation/physiology
8.
Pflugers Arch ; 467(4): 817-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24903240

ABSTRACT

The purpose of this study was to assess, in the murine kidney, the mechanisms underlying the endothelium-dependent control of vascular tone and whether or not, in a severe model of hypertension and renal failure, KCa channels contribute to its regulation. Wild-type (BL) and double-transgenic female mice expressing human angiotensinogen and renin (AR) genes received either control or a high-salt diet associated to a nitric oxide (NO) synthase inhibitor treatment (BLSL and ARSL). Changes in renal perfusion pressure (RPP) were measured in isolated perfused kidneys. BLSL and AR were moderately hypertensive without kidney disease while ARSL developed severe hypertension and renal failure. In the four groups, methacholine induced biphasic endothelium-dependent responses, a transient decrease in RPP followed by a cyclooxygenase-dependent increase in RPP. In the presence or not of indomethacin, the vasodilatations were poorly sensitive to NO synthase inhibition. However, in the presence of cyclooxygenase and NO synthase inhibitors, apamin, and/or TRAM-34, blockers of KCa2.3 and KCa3.1, respectively, abolished the decrease in RPP in response to either methacholine or the two activators of KCa2.3/KCa3.1, NS309, and SKA-31. Thus, KCa2/3 channels play a major role in the regulation of murine kidney perfusion and this mechanism is maintained in hypertension, even when severe and associated with kidney damage.


Subject(s)
Hypertension, Renovascular/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Renal Insufficiency/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Vasodilation , Angiotensinogen/genetics , Angiotensinogen/metabolism , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Female , Humans , Hypertension, Renovascular/etiology , Hypertension, Renovascular/physiopathology , Indomethacin/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Methacholine Chloride/pharmacology , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Renal Insufficiency/etiology , Renal Insufficiency/physiopathology , Renin/genetics , Renin/metabolism , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Sodium, Dietary/adverse effects
9.
J Vasc Res ; 49(4): 309-18, 2012.
Article in English | MEDLINE | ID: mdl-22572574

ABSTRACT

INTRODUCTION: Age and hypertension are two major determinants of arterial stiffness, as well as endothelial dysfunction. The present study was designed to test whether a chronic reduction of endogenous nitric oxide (NO) produces arterial stiffening close to that observed in old spontaneously hypertensive rats (SHR), and also to study the effect of an acute or a chronic decrease in blood pressure (BP) on aortic distensibility. METHODS: BP, aortic stiffness, endothelial dysfunction and remodelling were measured in male adult (20-week-old) SHR, in adult SHR treated with a nonspecific NO synthase inhibitor L-NAME (SHR/L-NAME) for 2 weeks, in adult SHR/L-NAME cotreated with perindopril (1 mg/kg/day) and in old SHR (55-week-old). Age-matched WKY were used as a normotensive group. RESULTS: Aortic endothelial dysfunction, remodelling and stiffening appeared in old SHR. Reduction of NO production in adult SHR caused similar alterations. Acute decreases in BP in SHR/L-NAME did not improve isobaric aortic distensibility but a chronic reduction of BP prevented endothelial dysfunction, aortic remodelling and aortic wall stiffening. CONCLUSION: NO reduction in adult SHR induces aortic alterations similar to those observed during aging, which supports the major role of NO in the development of arterial stiffening. These aortic alterations can be prevented by angiotensin-converting enzyme inhibitor treatment.


Subject(s)
Aging/pathology , Aorta/physiopathology , Hypertension/physiopathology , Nitric Oxide/antagonists & inhibitors , Vascular Stiffness/drug effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Aorta/drug effects , Blood Pressure/drug effects , Clonidine/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Perindopril/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY
10.
Am J Physiol Renal Physiol ; 300(6): F1310-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21411480

ABSTRACT

The (pro)renin receptor (PRR) has recently been demonstrated to bind equally well renin and its precursor, prorenin, leading to a similar intracellular signaling independent of angiotensin II. In this study, we report that human embryonic kidney cells (HEK) exposed to renin or prorenin for 24 h in the presence of a blocking concentration of the angtiotensin-converting enzyme inhibitor perindoprilate increased superoxide anion production as measured by luminescence (lucigenin) and electron spin resonance spectroscopy (hydroxylamine radical transition). Also, both renin and prorenin increased Nox4 expression while Nox2, p47(phox), and p67(phox) remained unchanged. In an investigation of the effects of renin and prorenin on fibrosis genes, it appeared that both proteins stimulated transforming growth factor-ß (TGF-ß), fibronectin, and plasminogen activator inhibitor type 1 (PAI-1) expression and therefore participated to an overall switch toward a profibrotic state of the kidney cells. When the cells were transfected with a siRNA targeting the PRR, Nox4 expression was efficiently prevented as well as the increase in superoxide production, TGF-ß, fibronectin, and PAI-1. Finally, we demonstrated that transfection of the cells with a Nox4-specific small interfering (si) RNA also prevented fibrosis gene expression following treatment with renin or prorenin. The results demonstrate that renin and prorenin, through their specific membrane receptor and independently of angiotensin II, promote fibrosis gene expression via a Nox4-dependent mechanism.


Subject(s)
Fibrosis/metabolism , Receptors, Cell Surface/metabolism , Renin/pharmacology , Superoxides/metabolism , Analysis of Variance , Blotting, Western , Cells, Cultured , Fibrosis/genetics , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Prorenin Receptor
11.
Thromb Res ; 124(4): 439-46, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19540572

ABSTRACT

INTRODUCTION: Plasminogen Activator Inhibitor-1 (PAI-1) is the most potent endogenous inhibitor of fibrinolysis which is implicated in the pathogenesis of myocardial infarction and metabolic syndrome. The formation of reactive oxygen species (ROS) plays an important role in the pathology of vascular disorders and has been shown to increase PAI-1 expression by endothelial cells. Growing evidence indicates that NADPH oxidase and in particular the constitutively active Nox4-p22(phox) complexes are major sources of ROS in endothelial cells. The aim of the present study was to characterize the role of NADPH oxidase and in particular Nox4 in the regulation of PAI-1 expression in cultured Human Umbilical Venous Endothelial Cells (HUVECs). METHODS AND RESULTS: N-acetylcysteine (NAC, scavenger of ROS), diphenylene iodonium chloride (DPI, inhibitor of flavoproteins), M40403 (superoxyde dismutase mimic) and S17834 (inhibitor of NADPH oxidase) inhibited PAI-1 release and promoter activity in HUVECs. Specific knock down of Nox4 mRNA by siRNA caused a decrease in ROS production and NADPH oxidase activity. Moreover, Nox4 silencing decreased PAI-1 expression, release and activity as well as p38 MAPK pathways and NFkappaB activation. These signalling pathways are also involved in PAI-1 release. CONCLUSIONS: The NADPH oxidase inhibitors DPI and S 17834 as well as Nox4 silencing decreased PAI-1 synthesis in human cultured endothelial cells demonstrating the involvement of the constitutively active Nox4-containing NADPH oxidase in ROS-mediated PAI-1 transcription via p38 MAPK pathways. NADPH oxidase targeting with inhibitors such as S17834 could be an interesting strategy to decrease both oxidative stress and PAI-1 synthesis.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , MAP Kinase Signaling System , NADPH Oxidases/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Benzopyrans/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Humans , Manganese , NADPH Oxidase 4 , NADPH Oxidases/genetics , Onium Compounds/pharmacology , Organometallic Compounds/pharmacology , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism
12.
J Cardiovasc Pharmacol ; 51(2): 131-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18287880

ABSTRACT

Human saphenous veins (SV) are used for coronary bypass surgery despite the higher rate of graft failure observed as compared to arteries. A higher production of reactive oxygen species (ROS) in SV than in internal mammary artery (IMA) has been incriminated as possibly implicated in graft failure. NADPH oxidase, involved in vascular ROS production, was therefore characterized in human smooth muscle cells from SV. ROS production was confirmed to be essentially NADPH oxidase dependent in cultured smooth muscle cells (SMC) from human SV and increased in comparison with IMA. To investigate the role of NADPH oxidase subunits, siRNA for nox1, nox2, or p47 mRNA were studied. In cultured venous SMC under unstimulated conditions, inhibition of nox1 or nox2 mRNA decreased ROS production, whereas p47 silencing increased it. During angiotensin II (AngII) activation, nox2 or p47 mRNA silencing decreased ROS production, while nox1 inhibition had no effect. Venous SMC express functional nox1 and nox2. Only nox2 is implicated in response to AngII whilst nox1 is involved in unstimulated ROS production. p47 negatively regulates ROS generation under basal conditions, whereas it enhances AngII increased ROS production. Thus, nox1, nox2, and p47 have distinct roles in NADPH oxidase activity in human veins.


Subject(s)
Angiotensin II/pharmacology , Muscle, Smooth, Vascular/drug effects , NADPH Oxidases/metabolism , Saphenous Vein , Aged , Aged, 80 and over , Analysis of Variance , Blotting, Western , Cells, Cultured , Enzyme Activation , Female , Humans , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Am J Physiol Renal Physiol ; 294(1): F120-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17942572

ABSTRACT

Early manifestations of kidney disease occur in atherosclerosis and activation of TP (thromboxane A(2)) receptors is implicated in atherosclerotic, diabetes, and renal diseases. The purpose of the present study was to analyze, in isolated, perfused mouse kidneys, the participation of TP receptors in renal vasoconstrictions and vasodilatations. In kidneys, taken from wild-type C57BL6, apolipoprotein E-deficient (ApoE-KO) and diabetic ApoE-KO mice, changes in perfusion pressure were recorded. Constrictions to TP receptor ligands U 46619, arachidonic acid, PGH(2), and 8-iso-PGF(2alpha), but not those to angiotensin II, endothelin, or norepinephrine, were inhibited by the selective TP receptor antagonist Triplion (S 18886; 10 nM). Acetylcholine and prostacyclin evoked biphasic responses during methoxamine constrictions; the constrictor part was blocked by Triplion. In ApoE-KO mouse kidneys, compared with C57BL6, a specific decrease in norepinephrine response and no modification in dilator responses were observed. In diabetic ApoE-KO mouse kidneys, constrictions to U 46619 and those to 8-iso-PGF(2alpha) were significantly and selectively augmented, without modification in the expression of the TP receptor, and again without any significant change in vasodilator activity. Thus TP receptors are functional, and their activation is not involved in norepinephrine, endothelin, and angiotensin II vasoconstrictions but is implicated in the unusual vasoconstrictions to acetylcholine and prostacyclin. Increased responsiveness of TP receptors occurs in diabetic ApoE-KO mouse kidneys. Thus early changes in TP receptor-mediated vasoconstrictor activity may participate in the development of kidney disease in atherosclerosis and diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Kidney/blood supply , Kidney/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Vasoconstriction/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Acetylcholine/pharmacology , Animals , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Dinoprost/analogs & derivatives , Dinoprost/pharmacology , Disease Models, Animal , Epoprostenol/pharmacology , Male , Methoxamine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthalenes/pharmacology , Propionates/pharmacology , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Receptors, Thromboxane A2, Prostaglandin H2/drug effects , Streptozocin , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/pharmacology
14.
Clin Sci (Lond) ; 112(4): 229-39, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17020541

ABSTRACT

Varicose vein disease is a frequently occurring pathology with multifactorial causes and a genetic component. An intense remodelling of the varicose vein wall has been described and could be at the origin of its weakness and altered elasticity. We have described previously a dysregulation of collagen synthesis in cultured smooth muscle cells from saphenous veins and in dermal fibroblasts from the skin of patients with varicose veins, suggesting a systemic defect in their connective tissue. The present study describes comparative morphological and immunohistochemical data in both the skin and saphenous veins of eight control subjects (undergoing coronary bypass surgery) and eight patients with varicose veins. Histological staining of glycoproteins, the elastic fibre network and collagen bundles showed that the remodelling and fragmentation of elastic fibres observed in varicose veins were also present in the skin of the patients. When compared with control subjects, we observed in both the veins and skin of patients with varicose veins (i) an increase in the elastic network, as quantified by image analysis; (ii) an accumulation of collagen type I, fibrillin-1 and laminin; and (iii) an overproduction of MMP (matrix metalloproteinase)-1, MMP-2 and MMP-3, analysed by immunohistochemistry, but normal levels of other MMPs (MMP-7 and MMP-9) and their inhibitors (TIMP-1, TIMP-2 and TIMP-3). An imbalance of extracellular matrix production/degradation was thus observed in veins as well as in the skin of the patients with varicose veins and, taken together, these findings show that remodelling is present in different organs, confirming systemic alterations of connective tissues.


Subject(s)
Extracellular Matrix/pathology , Saphenous Vein/pathology , Skin/pathology , Varicose Veins/pathology , Adult , Aged , Collagen Type I/metabolism , Extracellular Matrix/metabolism , Female , Fibrillin-1 , Fibrillins , Humans , Image Processing, Computer-Assisted/methods , Laminin/metabolism , Male , Matrix Metalloproteinases/biosynthesis , Microfilament Proteins/metabolism , Middle Aged , Saphenous Vein/metabolism , Skin/metabolism , Tissue Inhibitor of Metalloproteinases/biosynthesis , Varicose Veins/metabolism
15.
J Vasc Res ; 42(5): 388-98, 2005.
Article in English | MEDLINE | ID: mdl-16088212

ABSTRACT

An alteration of extracellular matrix is involved in varicose veins. We have previously shown that collagen III production, but not its mRNA expression, is decreased in cultured smooth muscle cells (SMC) from varicose veins, involving an over-production of collagen I. In this study, the mechanisms involved in this collagen III reduction are explored. Steady state levels of collagen III mRNA and its ability to translate a protein were evaluated. Neither stability nor functionality of the alpha1(III) coding mRNA were affected in cells from varicose veins. Potential intracellular degradations of collagen III were investigated with inhibitors of intracellular proteases but the production was unaffected. The level of N-terminal propeptides of collagen III in the extracellular medium was determined and was similar in SMC from control and varicose veins. The stability of collagen III was determined by time-course experiments and a degradation of the protein was observed in cells from varicose veins. The production of collagen III was partially restored in cells from varicose veins in the presence of Marimastat, a matrix metalloproteinase (MMP) inhibitor. The mRNA expression and protein production of MMP3 were increased in cells from varicose veins. Fibronectin, a potential substrate of MMP3, was decreased in SMC from varicose veins. In conclusion, collagen III, and probably fibronectin, are degraded extracellularly in SMC from varicose veins by a mechanism involving MMPs, and maybe MMP3 by a direct or an indirect pathway. The degradation of collagen III and fibronectin may have repercussions for the mechanical properties of the venous wall.


Subject(s)
Collagen Type III/genetics , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Muscle, Smooth, Vascular/physiopathology , Varicose Veins/physiopathology , Aged , Aged, 80 and over , Cell Compartmentation , Cells, Cultured , Collagen Type III/metabolism , Extracellular Space/metabolism , Female , Fibronectins/metabolism , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Procollagen/metabolism , Procollagen N-Endopeptidase/metabolism , Protease Inhibitors/metabolism , Protein Biosynthesis , RNA Stability , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Varicose Veins/metabolism , Varicose Veins/pathology
16.
Cardiovasc Res ; 63(2): 323-30, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15249190

ABSTRACT

OBJECTIVE: Anoxia followed by reoxygenation (A/R) increases endothelial cell superoxide (O2-) generation which is implicated in E-selectin overexpression. The mechanisms which govern these processes are not fully understood and therefore the goal of our study was to determine the functional importance of NADPH oxidase in the regulation of E-selectin expression in human umbilical veins endothelial cells (HUVECs) submitted to A/R. METHODS: O2- production was estimated using lucigenin chemiluminescence and formazan accumulation. NADPH oxidase expression in HUVECs was studied by RT-PCR and Western blot and E-selectin by Northern blot analysis. NFkappaB activation was assessed by electrophoretic mobility shift assay. RESULTS: A/R caused an increased O2- production which was inhibited by the superoxide dismutase mimetic M40403 (50 micromol/l), the protein kinase C inhibitor chelerythrine (10 micromol/l), the NADPH oxidase inhibitor diphenyleneiodonium (DPI, 10 micromol/l) and the NADPH oxidase assembly blocker apocynin (600 micromol/l). At the end of the anoxic period, the mRNA expression and the protein p47phox was increased as compared to normoxic HUVECs. NFkappaB activation of anoxic HUVECs was maximal after 1 h of reoxygenation and returned to basal normoxic levels after 2 h of reoxygenation. Apocynin reduced the NFkappaB activation at 1 h of reoxygenation. E-selectin mRNA expression was increased after 3 h of reoxygenation of anoxic HUVECs and the SOD mimetic M40403 as well as apocynin prevented this overexpression. CONCLUSIONS: Activated NADPH oxidase is a critical enzyme in E-selectin overexpression after A/R of HUVECs. Moreover, A/R increased expression of membranous and cytosolic NADPH oxidase subunits as well as the protein p47phox. Strategies aimed at preventing endothelial NADPH oxidase activation and/or activity may be useful in controlling leukocyte adhesion during ischemia/reperfusion.


Subject(s)
E-Selectin/metabolism , Endothelial Cells/metabolism , Hypoxia/metabolism , NADPH Oxidases/metabolism , Superoxides/metabolism , Blotting, Southern/methods , Blotting, Western/methods , Cells, Cultured , Humans , Membrane Transport Proteins/genetics , NADPH Dehydrogenase/genetics , Phosphoproteins/analysis , Phosphoproteins/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
17.
Angiology ; 54 Suppl 1: S13-8, 2003.
Article in English | MEDLINE | ID: mdl-12934753

ABSTRACT

Varicose vein disease is a common condition. Its pathology is not well characterized. A disorganization of smooth muscle cells and extracellular matrix components in the venous wall have been described. The objective of this paper is to offer an explanation for the abnormal distensibility of varicose veins. The content of hydroxyproline was quantified in control and varicose human saphenous veins. The synthesis of collagen types I, III, and V was quantified in cultured venous smooth muscle cells and dermal fibroblasts of control subjects and patients with varicose veins. The proportion of collagen type III in the heterofibrils composed by collagen types I, III, and V was calculated. The level of hydroxyproline was increased in varicose veins, suggesting an increased content of collagen. This augmentation of collagen in diseased tissues appears to be correlated with an increase of collagen type I since the collagen I mRNA was overexpressed in varicose veins, whereas collagen type III mRNA was not altered. The quantification of collagen synthesis in cultured cells shows that proportion of collagen type III was significantly decreased in cultured smooth muscle cells and dermal fibroblasts derived from patients with varicose veins. The results indicate a deficiency in collagen type III in patients with varicose veins. Since collagen type III is involved in tissue elasticity, these results offer an explanation for the abnormal distensibility of varicose veins. Moreover, this defect seems to be generalized in different tissues and argues in favor of a genetic alteration of remodeling in these patients.


Subject(s)
Collagen/biosynthesis , Varicose Veins/metabolism , Aged , Cells, Cultured , Collagen Type I/biosynthesis , Collagen Type III/biosynthesis , Collagen Type III/deficiency , Collagen Type V/biosynthesis , Female , Fibroblasts/metabolism , Humans , Hydroxyproline/metabolism , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , RNA, Messenger/genetics , Saphenous Vein/metabolism , Saphenous Vein/pathology
18.
Circulation ; 106(4): 479-83, 2002 Jul 23.
Article in English | MEDLINE | ID: mdl-12135949

ABSTRACT

BACKGROUND: The dilatation and tortuosity observed in varicose veins provide evidence for progressive venous wall remodeling associated with abnormalities of smooth muscle cells and extracellular matrix. The present study was designed to examine if the phenotypic modulations observed in the venous smooth muscle cells of patients with varicose veins were also present in their dermal fibroblasts. METHODS AND RESULTS: Collagen type I (collagen I), type III (collagen III), and type V (collagen V) were compared in dermal fibroblasts derived from the skin of control subjects and patients with varicose veins. The synthesis of collagen I, the release of its metabolites, and the expression of its mRNA were increased in fibroblasts from patients with varicose veins, whereas the synthesis of collagen III was decreased but not correlated with a decrease in mRNA expression and in metabolite release. Matrix metalloproteinases (MMP1, 2, 7, 8, 9, and 13) and their inhibitors (TIMP1 and 2) were quantified in both cell types; only the production of proMMP2 was increased in cells derived from patients with varicose veins. CONCLUSIONS: These findings suggest that the synthesis of collagen I and III is dysregulated in dermal fibroblasts derived from patients with varicose veins. These results are comparable with those observed in smooth muscle cells derived from varicose veins, thus suggesting a systemic alteration of tissue remodeling in subjects with varicose veins.


Subject(s)
Fibrillar Collagens/biosynthesis , Fibroblasts/metabolism , Skin/cytology , Varicose Veins/metabolism , Aged , Cell Division , Cells, Cultured , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/biosynthesis , Collagen Type III/genetics , Collagen Type III/metabolism , Culture Media/analysis , Female , Gene Expression Regulation , Humans , Kinetics , Male , Matrix Metalloproteinases/biosynthesis , Middle Aged , Muscle, Smooth, Vascular/metabolism , Protein Biosynthesis , RNA, Messenger/biosynthesis , Skin/metabolism , Tissue Inhibitor of Metalloproteinases/biosynthesis , Varicose Veins/genetics , Varicose Veins/pathology , Veins/cytology , Veins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...