Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Eur Respir J ; 34(5): 1100-10, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19324947

ABSTRACT

Mutations in the gene encoding bone morphogenetic protein (BMP) receptor type 2 (BMPR-2) have been reported in pulmonary arterial hypertension (PAH), but their functional relevance remains incompletely understood. BMP receptor expression was evaluated in human lungs and in cultured pulmonary artery smooth muscle cells (PASMCs) isolated from 19 idiopathic PAH patients and nine heritable PAH patients with demonstrated BMPR-2 mutations. BMP4-treated PASMCs were assessed for Smad and p38 mitogen-activated protein kinase (MAPK) signalling associated with mitosis and apoptosis. Lung tissue and PASMCs from heritable PAH patients presented with decreased BMPR-2 expression and variable increases in BMPR-1A and BMPR-1B expression, while a less important decreased BMPR-2 expression was observed in PASMCs from idiopathic PAH patients. Heritable PAH PASMCs showed no increased phosphorylation of Smad1/5/8 in the presence of BMP4, which actually activated the p38MAPK pathway. Individual responses varied from one mutation to another. PASMCs from PAH patients presented with an in vitro proliferative pattern, which could be inhibited by BMP4 in idiopathic PAH but not in heritable PAH. PASMCs from idiopathic PAH and more so from heritable PAH presented an inhibition of BMP4-induced apoptosis. Most heterogeneous BMPR-2 mutations are associated with defective Smad signalling compensated for by an activation of p38MAPK signalling, accounting for PASMC proliferation and deficient apoptosis.


Subject(s)
Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Adult , Apoptosis , Bone Morphogenetic Protein Receptors, Type II/genetics , Cell Proliferation , Endothelium, Vascular/pathology , Female , Hemodynamics , Humans , Male , Microcirculation , Mutation , Myocytes, Smooth Muscle/cytology , Pulmonary Artery/pathology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Eur Respir J ; 34(3): 731-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19324959

ABSTRACT

Common bile duct ligation (CBDL) induces biliary cirrhosis and pulmonary vasodilatation. We tested whether CBDL ameliorates monocrotaline (MCT)-induced pulmonary hypertension (PH) in rats. Five groups of rats were studied: controls; rats dosed with MCT (60 mg.kg(-1) subcutaneously); CBDL; rats dosed with MCT followed by CBDL on day 7; and rats dosed with MCT followed by CBDL (day 7) and L-NAME therapy between days 24 and 28. 28-day survival was 26% in the MCT group and 72% in the MCT+CBDL group. Pulmonary vascular resistance measured on days 21 and 28 increased in the MCT and MCT+CBDL+L-NAME groups, but returned to normal in the MCT+CBDL group on day 28. Pulmonary artery (PA) medial hypertrophy persisted in MCT+CBDL rats. PA inflammation increased in MCT+CBDL rats, with accumulation of both intra- and perivascular macrophages. Exhaled nitric oxide (NO) levels decreased in the MCT group and increased in the MCT+CBDL group, which showed upregulation of inducible NO synthase and normal endothelial NO synthase. Blood endothelin (ET)-1 increased in CBDL, MCT, and MCT+CBDL rats. Levels of ET(B) receptors increased and ET(A) receptors decreased in the MCT+CBDL group, whereas the opposite changes occurred in the MCT group. Biliary cirrhosis induces pulmonary vasodilation that ameliorates MCT-induced PH and improves survival. Upregulation of inducible NO synthase and ET(B) receptor and downregulation of ET(A) receptor may be involved.


Subject(s)
Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/prevention & control , Liver Cirrhosis, Biliary/etiology , Liver Cirrhosis, Biliary/physiopathology , Monocrotaline , Animals , Common Bile Duct , Disease Models, Animal , Endothelins/metabolism , Enzyme Inhibitors/therapeutic use , Hypertension, Pulmonary/metabolism , Ligation , Liver Cirrhosis, Biliary/metabolism , Male , NG-Nitroarginine Methyl Ester/therapeutic use , Nitric Oxide Synthase/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Vasodilation/physiology
3.
Circulation ; 113(1): 81-9, 2006 Jan 03.
Article in English | MEDLINE | ID: mdl-16380550

ABSTRACT

BACKGROUND: Serotonin (5-hydroxytryptamine; 5-HT) overproduction is responsible for cardiac valvular disease in patients with carcinoid tumors. Reduced 5-HT inactivation is one proposed mechanism of the valvulopathy observed in individuals treated with the appetite suppressants fenfluramine and phentermine. One key protein limiting systemic availability of 5-HT is the 5-HT transporter (5-HTT) expressed by platelets and pulmonary vascular cells; 5-HTT is responsible for 5-HT uptake and subsequent inactivation of the amine passing through the lung. Here we investigated whether 5-HTT-deficient (5-HTT-KO) mice developed structural and/or functional cardiac abnormalities and valvulopathy. METHODS AND RESULTS: Cardiac endothelial cells expressed large amounts of 5-HTT in wild-type mice. 5-HTT deficiency appeared to be associated with marked interstitial, perivascular, and valvular fibrosis as evidenced by staining of cardiac collagen in 5-HTT-KO mice. Histological analysis provided evidence for valvulopathy characterized by valvular hyperplasia and prominent fibrosis at the attachment site and base of the leaflets. Echocardiography revealed an increase in left ventricular lumen diameter and a decrease in left ventricular diameter fractional shortening. Although 5-HT1B receptors mediated the 5-HT-induced collagen secretion by human cardiac myofibroblasts, the contribution of this receptor type to valvulopathy was ruled out because double-KO mice deficient in both 5-HTT and 5-HT1B receptors showed the same cardiac alterations as 5-HTT-KO mice. CONCLUSIONS: The present results establish a link between 5-HTT and the development of cardiac fibrosis and valvulopathy in vivo. 5-HTT-KO mice represent an especially relevant model for studying the mechanisms by which 5-HT induces valvulopathy.


Subject(s)
Fibrosis/etiology , Heart Valve Diseases/etiology , Serotonin Plasma Membrane Transport Proteins/genetics , Animals , Fibroblasts/cytology , Fibrosis/pathology , Heart Valve Diseases/diagnostic imaging , Heart Valve Diseases/pathology , Humans , Hydroxyindoleacetic Acid/blood , Male , Mice , Mice, Knockout , Myocardium/pathology , RNA, Messenger/analysis , Receptor, Serotonin, 5-HT1B/genetics , Serotonin/blood , Serotonin Plasma Membrane Transport Proteins/deficiency , Serotonin Plasma Membrane Transport Proteins/physiology , Ultrasonography
4.
J Clin Invest ; 87(1): 155-62, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1985092

ABSTRACT

To determine whether exposure to chronic hypoxia and subsequent development of pulmonary hypertension induces alterations of endothelium-dependent relaxation in rat pulmonary vascular bed, we studied isolated lung preparations from rats exposed to either room air (controls) or hypoxia (H) during 1 wk (1W-H), 3 wk (3W-H), or 3W-H followed by 48 h recovery to room air (3WH + R). In lungs pretreated with meclofenamate (3 microM), the endothelium-dependent vasodilator responses to acetylcholine (10(-9)-10(-6) M) and ionophore A23187 (10(-9)-10(-7) M) were examined during conditions of increased tone by U46619 (50 pmol/min). Acetylcholine or A23187 produced dose-dependent vasodilation in control lungs, this response was reduced in group 1W-H (P less than 0.02), abolished in group 3W-H (P less than 0.001), and restored in group 3WH + R. In contrast, the endothelium-independent vasodilator agent sodium nitroprusside remained fully active in group 3W-H. The pressor response to 300 pM endothelin was greater in group 3W-H than in controls (6.8 +/- 0.5 mmHg vs. 1.6 +/- 0.2 mmHg, P less than 0.001) but was not potentiated by the endothelium-dependent relaxing factor (EDRF) antagonists: hydroquinone (10(-4) M); methylene blue (10(-4) M); and pyrogallol (3 x 10(-5) M) as it was in controls. It was similar to controls in group 3W-H + R. Our results demonstrate that hypoxia-induced pulmonary hypertension is associated with a loss of EDRF activity in pulmonary vessels, with a rapid recovery on return to a normoxic environment.


Subject(s)
Hypoxia/physiopathology , Nitric Oxide/physiology , Pulmonary Circulation , Acetylcholine/pharmacology , Animals , Blood Pressure/drug effects , Calcimycin/pharmacology , Chronic Disease , Endothelins/pharmacology , Endothelium, Vascular/physiology , Hypertension, Pulmonary/physiopathology , Male , Nitroprusside/pharmacology , Rats , Rats, Inbred Strains
5.
J Clin Invest ; 94(2): 578-84, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8040313

ABSTRACT

Exposure to hypoxia and subsequent development of pulmonary hypertension is associated with an impairment of the nitric oxide (NO) mediated response to endothelium-dependent vasodilators. Inhaled NO may reach resistive pulmonary vessels through an abluminal route. The aim of this study was to investigate if continuous inhalation of NO would attenuate the development of pulmonary hypertension in rats exposed to chronic hypoxia. In conscious rats previously exposed to 10% O2 for 3 wk, short-term inhalation of NO caused a dose-dependent decrease in pulmonary artery pressure (PAP) from 44 +/- 1 to 32 +/- 1 mmHg at 40 ppm with no changes in systemic arterial pressure, cardiac output, or heart rate. In normoxic rats, acute NO inhalation did not cause changes in PAP. In rats simultaneously exposed to 10% O2 and 10 ppm NO during 2 wk, right ventricular hypertrophy was less severe (P < 0.01), and the degree of muscularization of pulmonary vessels at both alveolar duct and alveolar wall levels was lower (P < 0.01) than in rats exposed to hypoxia alone. Tolerance to the pulmonary vasodilator effect of NO did not develop after prolonged inhalation. Brief discontinuation of NO after 2 wk of hypoxia plus NO caused a rapid increase in PAP. These data demonstrate that prolonged inhalation of low concentrations of NO induces sustained pulmonary vasodilation and reduces pulmonary vascular remodeling in response to chronic hypoxia.


Subject(s)
Hypertension, Pulmonary/prevention & control , Hypoxia/complications , Nitric Oxide/pharmacology , Administration, Inhalation , Animals , Chronic Disease , Hypertension, Pulmonary/etiology , Male , Nitric Oxide/administration & dosage , Pulmonary Artery/physiology , Rats , Rats, Wistar
6.
J Clin Invest ; 105(11): 1555-62, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10841514

ABSTRACT

Hypoxia is a well-recognized stimulus for pulmonary blood vessel remodeling and pulmonary hypertension development. One mechanism that may account for these effects is the direct action of hypoxia on the expression of specific genes involved in vascular smooth muscle cell (SMC) proliferation. Previous studies demonstrated that the serotonin (5-hydroxytryptamine; 5-HT) transporter (5-HTT) mediates the mitogenic activity of 5-HT in pulmonary vascular SMCs and is overexpressed during hypoxia. Thus, 5-HT-related mitogenic activity is increased during hypoxia. Here, we report that mice deficient for 5-HTT (5-HTT(-/-)) developed less hypoxic pulmonary hypertension and vascular remodeling than paired 5-HTT(+/+) controls. When maintained under normoxia, 5-HTT(-/-)-mutant mice had normal hemodynamic parameters, low blood 5-HT levels, deficient platelet 5-HT uptake, and unchanged blood levels of 5-hydroxyindoleacetic acid, a metabolite of 5-HT. After exposure to 10% O(2) for 2 or 5 weeks, the number and medial wall thickness of muscular pulmonary vessels were reduced in hypoxic 5-HTT(-/-) mice as compared with wild-type paired controls. Concomitantly, right ventricular systolic pressure was lower and right ventricle hypertrophy less marked in the mutant mice. This occurred despite potentiation of acute hypoxic pulmonary vasoconstriction in the 5-HTT(-/-) mice. These data further support a key role of 5-HTT in hypoxia-induced pulmonary vascular SMC proliferation and pulmonary hypertension.


Subject(s)
Carrier Proteins/physiology , Hypertension, Pulmonary/etiology , Hypoxia/physiopathology , Membrane Glycoproteins/physiology , Membrane Transport Proteins , Nerve Tissue Proteins , Animals , Carrier Proteins/analysis , Carrier Proteins/genetics , Hypertension, Pulmonary/prevention & control , Immunohistochemistry , Lung/chemistry , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins
7.
J Clin Invest ; 108(8): 1141-50, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11602621

ABSTRACT

Hyperplasia of pulmonary artery smooth muscle cells (PA-SMCs) is a hallmark pathological feature of primary pulmonary hypertension (PPH). Here we found that PA-SMCs from patients with PPH grow faster than PA-SMCs from controls when stimulated by serotonin or serum and that these effects are due to increased expression of the serotonin transporter (5-HTT), which mediates internalization of indoleamine. In the presence of 5-HTT inhibitors, the growth stimulatory effects of serum and serotonin were markedly reduced and the difference between growth of PA-SMCs from patients and controls was no longer observed. As compared with controls, the expression of 5-HTT was increased in cultured PA-SMCs as well as in platelets and lungs from patients with PPH where it predominated in the media of thickened pulmonary arteries and in onion-bulb lesions. The L-allelic variant of the 5HTT gene promoter, which is associated with 5-HTT overexpression and increased PA-SMC growth, was present in homozygous form in 65% of patients but in only 27% of controls. We conclude that 5-HTT activity plays a key role in the pathogenesis of PA-SMC proliferation in PPH and that a 5HTT polymorphism confers susceptibility to PPH.


Subject(s)
Carrier Proteins/genetics , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Membrane Glycoproteins/genetics , Membrane Transport Proteins , Muscle, Smooth, Vascular/pathology , Nerve Tissue Proteins , Pulmonary Artery/pathology , Adolescent , Adult , Aged , Alleles , Carrier Proteins/blood , Carrier Proteins/metabolism , Case-Control Studies , Cells, Cultured , Female , Gene Expression , Humans , Hyperplasia , Hypertension, Pulmonary/metabolism , Lung/metabolism , Male , Membrane Glycoproteins/metabolism , Middle Aged , Serotonin Plasma Membrane Transport Proteins , Thymidine/metabolism
8.
Int J Clin Pract Suppl ; (158): 19-25, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18078390

ABSTRACT

Pulmonary artery hypertension (PAH) is now considered to be a proliferative disorder characterised by unexplained proliferation of pulmonary artery smooth muscle cells (PA-SMCs) and pulmonary artery endothelial cells (PA-ECs). An abnormal phenotype of PA-SMCs and PA-ECs has been described in PAH and some analogies now appear between pulmonary vascular cells from patients with PAH and cancer cells. Such analogies are discussed here with respect to essential hallmarks of cancer cells and with the hope that new treatments targeted at one or more of these cancer cell abnormalities may be appropriate for PAH.


Subject(s)
Hypertension, Pulmonary/pathology , Neoplasms/pathology , Apoptosis , Cell Division , Growth Inhibitors/physiology , Growth Substances/physiology , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/pathology , Neoplasms/metabolism
9.
Circ Res ; 87(5): 418-25, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10969041

ABSTRACT

Chronic hypoxic pulmonary hypertension (PH) results from persistent vasoconstriction, excess muscularization, and extracellular matrix remodeling of pulmonary arteries. The matrix metalloproteinases (MMPs) are a family of proteinases implicated in extracellular matrix turnover and hence in smooth muscle and endothelial cell migration and proliferation. Because MMP expression and activity are increased in PH, we designed the present study to investigate whether inhibition of lung MMPs in rats subjected to chronic hypoxia (CH) contributes to or protects against vascular remodeling and PH. To achieve lung MMP inhibition, rats exposed to 10% O(2) for 15 days were treated with either doxycycline (20 mg/kg per day by gavage starting 2 days before and continuing throughout the CH period) or a single dose of recombinant adenovirus (Ad) for the human tissue inhibitors of metalloproteinases-1 (hTIMP-1) gene (Ad.hTIMP-1, 10(8) plaque-forming units given intratracheally 2 days before CH initiation). Control groups either received no treatment or were treated with an adenovirus containing no gene in the expression cassette (Ad.Null). Efficacy of hTIMP-1 gene transfer was assessed both by ELISA on bronchoalveolar lavages and by hTIMP-1 immunofluorescence on lung sections. MMP inhibition in lungs was evaluated by in situ zymography and gelatinolytic activity assessment using [(3)H]gelatin. Rats treated with either doxycycline or Ad.hTIMP-1 had higher pulmonary artery pressure and right heart ventricular hypertrophy more severe than their respective controls. Worsening of PH was associated with increased muscularization and periadventitial collagen accumulation in distal arteries. In conclusion, our study provides compelling evidence that MMPs play a pivotal role in protecting against pulmonary artery remodeling.


Subject(s)
Genetic Therapy/methods , Hypertension, Pulmonary/drug therapy , Lung/enzymology , Matrix Metalloproteinase Inhibitors , Tissue Inhibitor of Metalloproteinase-1/therapeutic use , Tissue Inhibitor of Metalloproteinases/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Blood Pressure/drug effects , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Doxycycline/therapeutic use , Gelatinases/metabolism , Gene Transfer Techniques , Genetic Vectors , Hypertension, Pulmonary/etiology , Hypoxia , Immunohistochemistry , Lung/blood supply , Lung/chemistry , Matrix Metalloproteinases/biosynthesis , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Rats , Rats, Wistar , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics
10.
Arch Mal Coeur Vaiss ; 99(6): 621-5, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16878724

ABSTRACT

The nature of the primary defect responsible for triggering and maintaining pulmonary artery smooth muscle (PA-SMC) proliferation in pulmonary artery hypertension (PH) is poorly understood but may be either an inherent characteristic of PA-SMCs or a secondary response to an external abnormality, such as up-regulation of growth factors. In previous studies, we found that cultured PA-SMCs from patients with idiopathic PH (iPH) had an abnormally strong proliferative response to serotonin or serum (which contains high levels of serotonin). This abnormal response is due to overexpression of the serotonin transporter (5-HTT) which mediates the mitogenic action of serotonin. That 5-HTT plays a key role in pulmonary vascular remodeling is supported by experimental studies showing that transgenic animals overexpressing 5-HTT in smooth muscle (at a level close to that seen in PH) spontaneously develop pulmonary vascular remodeling and PH. Conversely, mice with targeted S-HTT gene disruption are protected against hypoxic PH, and selective 5-HTT inhibitors reverse or prevent experimental PH. In patients with chronic lung disease, a close association has been found between a 5-HTT gene polymorphism and the severity of pulmonary hypertension. Agents capable of selectively inhibiting 5-HTT-mediated PA-SMC proliferation deserve to be investigated as potential treatments for pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Animals , Humans , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Polymorphism, Genetic , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism
11.
Rev Mal Respir ; 23 Suppl 2: 4S45-4S51, 2006 Apr.
Article in French | MEDLINE | ID: mdl-16733400

ABSTRACT

INTRODUCTION: Pulmonary arterial hypertension (PAH) is related to hypertrophic remodelling of the pulmonary vessels. Proliferation of pulmonary vascular smooth muscle cells (PA-SMCs) plays a key role in these changes. BACKGROUND: The PA-SMCs of patients suffering from idiopathic or secondary PAH are characterized by abnormally increased in vitro proliferation in response to serotonin or serum. Serotonin transporter (5-HTT), the mediator of the mitogenic activity of serotonin, and the expression of which is increased in the course of idiopathic or secondary PAH, is the basis of these effects. The overexpression of 5-HTT, selectively induced in vascular smooth muscle by transgenesis in the mouse, leads to the development of PAH. Treatment with selective 5-HTT inhibitors prevents or leads to complete reversal of experimental hypoxic or inflammatory PAH. The presence of polymorphism of the gene causing over expression of the protein and proliferation of PA-SMCs seems to determine the severity of certain types of human PAH, notably that complicating COPD. VIEWPOINT: There is a potential therapeutic application of 5-HTT inhibitors in human PAH. A clinical study is currently taking place in France. CONCLUSION: The exploration of the role of 5-HTT and the mechanisms leading to its overexpression in PAH, as well as the interactions between 5-HTT and the BMPR2 gene, should lead to a significant increase in the understanding of the pathophysiology of the disease.


Subject(s)
Hypertension, Pulmonary/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/therapy , Muscle, Smooth, Vascular/metabolism , Polymorphism, Genetic , Pulmonary Disease, Chronic Obstructive/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Selective Serotonin Reuptake Inhibitors/therapeutic use , Severity of Illness Index
12.
PLoS One ; 10(6): e0131940, 2015.
Article in English | MEDLINE | ID: mdl-26121334

ABSTRACT

OBJECTIVE: Pulmonary artery smooth muscle cells (PA-SMCs) in pulmonary arterial hypertension (PAH) show similarities to cancer cells. Due to the growth-suppressive and pro-apoptotic effects of p53 and its inactivation in cancer, we hypothesized that the p53 pathway could be altered in PAH. We therefore explored the involvement of p53 in the monocrotaline (MCT) rat model of pulmonary hypertension (PH) and the pathophysiological consequences of p53 inactivation in response to animal treatment with pifithrin-α (PFT, an inhibitor of p53 activity). METHODS AND RESULTS: PH development was assessed by pulmonary arterial pressure, right ventricular hypertrophy and arterial wall thickness. The effect of MCT and PFT on lung p53 pathway expression was evaluated by western blot. Fourteen days of daily PFT treatment (2.2 mg/kg/day), similar to a single injection of MCT (60 mg/kg), induced PH and aggravated MCT-induced PH. In the first week after MCT administration and prior to PH development, p53, p21 and MDM2 protein levels were significantly reduced; whereas PFT administration effectively altered the protein level of p53 targets. Anti-apoptotic and pro-proliferative effects of PFT were revealed by TUNEL and MTT assays on cultured human PA-SMCs treated with 50 µM PFT. CONCLUSIONS: Pharmacological inactivation of p53 is sufficient to induce PH with a chronic treatment by PFT, an effect related to its anti-apoptotic and pro-proliferative properties. The p53 pathway was down-regulated during the first week in the rat MCT model. These in vivo experiments implicate the p53 pathway at the initiation stages of PH pathogenesis.


Subject(s)
Hypertension, Pulmonary/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Benzothiazoles/pharmacology , Cell Proliferation/drug effects , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertrophy, Right Ventricular/metabolism , Male , Monocrotaline/toxicity , Rats , Rats, Wistar , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/genetics
13.
Hum Gene Ther ; 12(5): 503-13, 2001 Mar 20.
Article in English | MEDLINE | ID: mdl-11268283

ABSTRACT

Endogenous as well as exogenous atrial natriuretic peptide (ANP) attenuates the development of chronic hypoxic pulmonary hypertension (CHPH) in rats. We built a recombinant adenovirus type 5 containing ANP cDNA under the control of the Rous sarcoma virus long terminal repeat (Ad.ANP). The efficiency of this vector in delivering the ANP gene was first examined in rat primary cultures of pulmonary vessel smooth muscle cells (SMCs) in comparison with Ad.beta GAL. Conditioned medium collected from Ad.ANP-infected cells (1000 TCID(50)/cell) contained 5 x 10(9) M immunoreactive ANP and elicited relaxation of isolated rat pulmonary arteries preconstricted with phenylepinephrine. To examine the effects of adenovirus-mediated ANP expression in the CHPH rat lung, Ad.ANP or Ad.beta GAL was administered via the tracheal route. Immunoreactive ANP was detected in bronchoalveolar fluid as early as 4 days and until 10-17 days after Ad.ANP administration (5 x 10(8) TCID(50)). Lung ANP immunostaining was mainly localized in bronchial and alveolar epithelial cells. As compared with Ad.beta GAL-treated controls, rats given Ad.ANP (5 x 10(8) TCID(50)) on the day before a 2-week exposure to hypoxia (10% O(2)) had lower values for pulmonary artery pressure (32.1 +/- 1.93 vs. 35.5 +/- 2 mmHg, p < 0.01) and Fulton's index (0.52 +/- 0.089 vs. 0.67 +/- 0.12, p < 0.001) and less severe right ventricular hypertrophy and distal vessel muscularization. These results suggest that induction of ANP expression in the lung may hold promise in the treatment of pulmonary hypertension.


Subject(s)
Adenoviridae/genetics , Atrial Natriuretic Factor/genetics , Hypertension, Pulmonary/prevention & control , Lung/metabolism , Animals , Atrial Natriuretic Factor/biosynthesis , Avian Sarcoma Viruses/genetics , Body Weight , Bronchoalveolar Lavage Fluid , Cells, Cultured , Culture Media, Conditioned , Cyclic GMP/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Epinephrine/pharmacology , Gene Transfer Techniques , Hypoxia , Immunohistochemistry , Muscle, Smooth/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors , Tissue Distribution , Trachea/metabolism , Transfection , Transgenes
14.
Br J Pharmacol ; 125(4): 681-8, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9831902

ABSTRACT

1. Cyclic guanosine 3'-5'-monophosphate (cyclic GMP) is the second messenger of important physiologically active mediators controlling the pulmonary vascular tone. To potentiate the effects of cyclic GMP on the pulmonary vasculature, we used DMPPO, a new selective PDE-5 inhibitor, and examined its action in a rat model of hypoxic pulmonary hypertension. 2. Levels of cyclic GMP measured during baseline conditions at 5 and 60 min of perfusion were similar in the perfusate of isolated lungs from normoxic and chronically hypoxic rats and did not differ with time. Pretreatment with DMPPO (1 microM) induced a larger increase in cyclic GMP concentration in the perfusate from chronically hypoxic rat lungs (31+/-36 at 5 min to 1821+/-83 pmol ml(-1) at 60 min) than in normoxic rat lungs (329+/-20 to 1281+/-127 pmol ml(-1), P<0.05). 3. In isolated lungs preconstricted with U-46619, pretreatment with DMPPO (1 microM) potentiated the vasodilator effects of atrial natriuretic peptide (100 pM-10 nM) and sodium nitroprusside (1 pM 10 nM), but did not alter vasodilation to isoproterenol. 4. In conscious rats previously exposed to 15 days hypoxia and studied under 10% O2, DMPPO (0.01, 0.05 and 0.1 mg kg(-1), i.v. bolus) caused a dose-dependent decrease in pulmonary arterial pressure (Pap) with no change in systemic artery pressure (Sap) and cardiac output. 5. Continuous infusion of DMPPO (0.1 mg kg(-1) h(-1) i.v. by osmotic pumps) in rats exposed to 10% O2 during 2-weeks reduced the Pap (P<0.05) and the degree of muscularization of pulmonary vessels at the alveolar wall (P<0.01) and alveolar duct levels (P<0.05) despite no significant change in right ventricular hypertrophy. 6. These results suggest that cyclic GMP phosphodiesterase inhibition may selectively dilate pulmonary circulation during chronic hypoxia.


Subject(s)
Allopurinol/analogs & derivatives , Cyclic GMP/pharmacology , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Phosphodiesterase Inhibitors/pharmacology , Vasodilation/drug effects , Allopurinol/pharmacology , Animals , Atrial Natriuretic Factor/pharmacology , Cyclic GMP/metabolism , Drug Interactions , Hypertrophy, Right Ventricular/physiopathology , In Vitro Techniques , Isoproterenol/pharmacology , Male , Myocardial Contraction/drug effects , Nitroprusside/pharmacology , Rats , Rats, Wistar , Time Factors
15.
J Appl Physiol (1985) ; 70(2): 567-74, 1991 Feb.
Article in English | MEDLINE | ID: mdl-2022547

ABSTRACT

This study investigated the pulmonary vascular response to endothelin (ET) in rats. In conscious rats, an incremental intravenous bolus of ET-1 (100-1,000 pM) caused, after an initial drop in systemic arterial pressure (Psa), a secondary dose-dependent increase of Psa concomitant with a decrease of cardiac output (CO) and heart rate (HR). Pulmonary arterial pressure (Ppa) remained unchanged, and pulmonary vascular resistance (PVR) increased significantly only after 1,000 pM (+ 40.0 +/- 10.4 at 15 min). Meclofenamate (6 mg/kg iv) did not alter hemodynamic response to ET (300 pM). After autonomic blockade with hexamethonium (6 mg/kg iv) plus atropine (0.75 mg/kg iv), bradycardia response to ET (300 pM) was blocked, but CO decreased, systemic vascular resistance increased, and PVR remained unchanged as in controls. In anesthetized ventilated rats, bolus injections of ET (10-1,000 pM) induced a transient dose-related decrease in compliance (-10.9 +/- 1.8% after 1,000 pM) but no change of conductance. In isolated lungs, Ppa increased at doses greater than 100 pM, and edema developed in response to 1,000 pM ET. The rise of Ppa in response to 300 pM was not altered by meclofenamate (3.2 x 10(-6) M) but was potentiated by inhibitors of endothelium-derived relaxing factor(s) (EDRF), methylene blue (10(-4) M), pyrogallol (3 x 10(-5) M), and NG-monomethyl-L-arginine (6 x 10(-4) M) (3.9 +/- 0.3, 4.6 +/- 0.5, and 5.9 +/- 0.3 mmHg, respectively, compared with 1.5 +/- 0.5 mmHg in control lungs). These results suggest that circulating ET is a more potent constrictor of the systemic circulation than of the pulmonary vascular bed.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Endothelins/pharmacology , Pulmonary Circulation/drug effects , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Endothelins/physiology , Heart Rate/drug effects , Heart Rate/physiology , In Vitro Techniques , Male , Nitric Oxide/metabolism , Pulmonary Circulation/physiology , Rats , Rats, Inbred Strains , Respiratory Mechanics/drug effects , Respiratory Mechanics/physiology , Vascular Resistance/drug effects , Vascular Resistance/physiology , Vasoconstriction/drug effects , Vasoconstriction/physiology
16.
Arch Mal Coeur Vaiss ; 87 Spec No 4: 41-51, 1994 Dec.
Article in French | MEDLINE | ID: mdl-7786135

ABSTRACT

Nitric oxide (NO) synthesised by endothelial cells, plays a key role in the control of vascular tone. Its synthesis from L-arginine is assured by NO-synthase, the activity of which is dependent on intracellular calcium concentrations, which are themselves modulated by pharmacological (acetylcholine, serotonin, bradykinin...) or physical factors (shearing forces exerted by blood flow). NO acts by stimulating a soluble guanylate-cyclase of the smooth muscle cells in the vessel wall. Its vasodilator effect is therefore mediated by an increase in intracellular cyclic GMP concentration. The synthesis or liberation of NO by the endothelium may be decreased or abolished during many pathological processes (hypercholesterolaemia, atherosclerosis, systemic or pulmonary hypertension...). The significance of this abnormality of NO-mediated endothelium-dependent vasodilation in different pathological conditions has not been established. However, it is probably significant in view of the different properties of NO: vaso-relaxation, antiaggregant and inhibition of vascular smooth muscle growth. It is not yet known whether this abnormality is a cause or a consequence of the underlying disease. From the therapeutic point of view, NO is an active metabolite of nitrate derivatives, sodium nitroprussiate and molsidomine which therefore share the same mode of action as the so-called "endothelium-dependent" vasodilatoe agents. The inhalation of NO, which is increasingly used in neonatal and adult intensive care units, is an alternative therapeutic approach in many conditions associated with pulmonary hypertension.


Subject(s)
Nitric Oxide/physiology , Administration, Inhalation , Animals , Endothelium, Vascular/physiology , Female , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Male , Nitric Oxide/administration & dosage , Nitric Oxide/therapeutic use , Vasodilation/physiology
20.
Am J Physiol Lung Cell Mol Physiol ; 293(4): L1045-52, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17675372

ABSTRACT

Serotonin [5-hydroxytryptamine (5-HT)] biosynthesis depends on two rate-limiting tryptophan hydroxylases (Tph): Tph1, which is expressed in peripheral organs, and Tph2, which is expressed in neurons. Because 5-HT is involved in pulmonary hypertension (PH), we investigated whether genetic variations in Tph1 and/or Tph2 affected PH development in mice. To examine the functional impact of peripheral Tph1 deficiency on hypoxic PH, we used Tph1(-/-) mice characterized by very low 5-HT synthesis rates and contents in the gut and lung and increased 5-HT synthesis in the forebrain. With chronic hypoxia, 5-HT synthesis in the forebrain increased further. Hypoxic PH, right ventricular hypertrophy, and distal pulmonary artery muscularization were less severe (P < 0.001) than in wild-type controls. The Tph inhibitor p-chlorophenylalanine (100 mgxkg(-1)xday(-1)) further improved these parameters. We then investigated whether mouse strains harboring the C1473G polymorphism of the Tph2 gene showed different PH phenotypes during hypoxia. Forebrain Tph activity was greater and hypoxic PH was more severe in C57Bl/6 and 129X1/SvJ mice homozygous for the 1473C allele than in DBA/2 and BALB/cJ mice homozygous for the 1473G allele. p-Chlorophenylalanine reduced PH in all groups and abolished the difference in PH severity across mouse strains. Hypoxia increased 5-hydroxytryptophan accumulation but decreased 5-HT contents in the forebrain and lung, suggesting accelerated 5-HT turnover during hypoxia. These results provide evidence that dysregulation of 5-HT synthesis is closely linked to the hypoxic PH phenotype in mice and that Tph1 and Tph2 may contribute to PH development.


Subject(s)
Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Polymorphism, Genetic , Tryptophan Hydroxylase/deficiency , Tryptophan Hydroxylase/genetics , Animals , Chronic Disease , Enzyme Inhibitors/pharmacology , Fenclonine/pharmacology , Hypertension, Pulmonary/genetics , Intestinal Mucosa/metabolism , Lung/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Prosencephalon/metabolism , Pulmonary Artery/physiopathology , Serotonin/biosynthesis , Serotonin/metabolism , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL