Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 317(6): L805-L815, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31577161

ABSTRACT

Histological observations in human pulmonary arterial hypertension (PAH) suggest a link between plexiform lesions and pulmonary supernumerary arteries. Pulmonary microvascular endothelial cells are characterized as hyperproliferative and progenitor-like. This study investigates the hypothesis that aneurysm-type plexiform lesions form in pulmonary supernumerary arteries because of their anatomical properties and endothelial characteristics similar to pulmonary microvascular endothelial cells. To induce PAH, rats were injected with Sugen5416, and exposed to hypoxia (10% O2) for 3 days (early stage) or 3 wk (mid-stage), or 3 wk of hypoxia with an additional 10 wk of normoxia (late-stage PAH). We examined morphology of pulmonary vasculature and vascular remodeling in lung serial sections from PAH and normal rats. Aneurysm-type plexiform lesions formed in small side branches of pulmonary arteries with morphological characteristics similar to supernumerary arteries. Over the course of PAH development, the number of Ki67-positive cells increased in small pulmonary arteries, including supernumerary arteries, whereas the number stayed consistently low in large pulmonary arteries. The increase in Ki67-positive cells was delayed in supernumerary arteries compared with small pulmonary arteries. In late-stage PAH, ~90% of small unconventional side branches that were likely to be supernumerary arteries were nearly closed. These results support our hypothesis that supernumerary arteries are the predominant site for aneurysm-type plexiform lesions in Sugen5416/hypoxia/normoxia-exposed PAH rats partly because of the combination of their unique anatomical properties and the hyperproliferative potential of endothelial cells. We propose that the delayed and extensive occlusive lesion formation in supernumerary arteries could be a preventive therapeutic target in patients with PAH.


Subject(s)
Aneurysm/pathology , Cell Proliferation , Disease Models, Animal , Pulmonary Arterial Hypertension/pathology , Pulmonary Arterial Hypertension/prevention & control , Pulmonary Artery/pathology , Vascular Remodeling , Aneurysm/etiology , Animals , Male , Pulmonary Arterial Hypertension/complications , Rats , Rats, Sprague-Dawley
2.
Am J Physiol Lung Cell Mol Physiol ; 311(4): L766-L769, 2016 10.
Article in English | MEDLINE | ID: mdl-27591244

ABSTRACT

Chronic thromboembolic pulmonary hypertension (CTEPH) is a hot topic in the field of pulmonary hypertension, because many CTEPH patients are now curable by surgical pulmonary endarterectomy and more recently possibly by pulmonary balloon angioplasty. However, there are still uncertainties regarding the pathogenesis of CTEPH, specifically how and where the small vessel arteriopathy that is indistinguishable from that in pulmonary arterial hypertension (plexogenic arteriopathy) develops, and how pulmonary endarterectomy improves hemodynamics and possibly cures CTEPH. Based on our recent experimental finding that hemodynamic stress is fundamental for the development of plexogenic arteriopathy, we discuss the uncertainties of CTEPH and potential implication of the effectiveness of pulmonary endarterectomy for reversing plexogenic arteriopathy and possibly providing a novel approach to cure pulmonary arterial hypertension.

3.
Am J Pathol ; 184(2): 369-75, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24401613

ABSTRACT

A major limitation in the pharmacological treatment of pulmonary arterial hypertension (PAH) is the lack of pulmonary vascular selectivity. Recent studies have identified a tissue-penetrating homing peptide, CARSKNKDC (CAR), which specifically homes to hypertensive pulmonary arteries but not to normal pulmonary vessels or other tissues. Some tissue-penetrating vascular homing peptides have a unique ability to facilitate transport of co-administered drugs into the targeted cells/tissues without requiring physical conjugation of the drug to the peptide (bystander effect). We tested the hypothesis that co-administered CAR would selectively enhance the pulmonary vascular effects of i.v. vasodilators in Sugen5416/hypoxia/normoxia-exposed PAH rats. Systemically administered CAR was predominantly detected in cells of remodeled pulmonary arteries. Intravenously co-administered CAR enhanced pulmonary, but not systemic, effects of the vasodilators, fasudil and imatinib, in PAH rats. CAR increased lung tissue imatinib concentration in isolated PAH lungs without increasing pulmonary vascular permeability. Sublingual CAR was also effective in selectively enhancing the pulmonary vasodilation by imatinib and sildenafil. Our results suggest a new paradigm in the treatment of PAH, using an i.v./sublingual tissue-penetrating homing peptide to selectively augment pulmonary vascular effects of nonselective drugs without the potentially problematic conjugation process. CAR may be particularly useful as an add-on therapy to selectively enhance the pulmonary vascular efficacy of any ongoing drug treatment in patients with PAH.


Subject(s)
Drug Delivery Systems/methods , Hypertension, Pulmonary/drug therapy , Peptides/chemistry , Vasodilator Agents/therapeutic use , Administration, Sublingual , Amino Acid Sequence , Animals , Arterial Occlusive Diseases/drug therapy , Arterial Occlusive Diseases/pathology , Benzamides/pharmacology , Benzamides/therapeutic use , Familial Primary Pulmonary Hypertension , Hemodynamics/drug effects , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Imatinib Mesylate , Infusions, Intravenous , Injections, Intravenous , Male , Molecular Sequence Data , Piperazines/pharmacology , Piperazines/therapeutic use , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Treatment Outcome , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacology
4.
Am J Physiol Heart Circ Physiol ; 306(2): H243-50, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24240870

ABSTRACT

We have investigated the temporal relationship between the hemodynamic and histological/morphological progression in a rat model of pulmonary arterial hypertension that develops pulmonary arterial lesions morphologically indistinguishable from those in human pulmonary arterial hypertension. Adult male rats were injected with Sugen5416 and exposed to hypoxia for 3 wk followed by a return to normoxia for various additional weeks. At 1, 3, 5, 8, and 13 wk after the Sugen5416 injection, hemodynamic and histological examinations were performed. Right ventricular systolic pressure reached its maximum 5 wk after Sugen5416 injection and plateaued thereafter. Cardiac index decreased at the 3∼5-wk time point, and tended to further decline at later time points. Reflecting these changes, calculated total pulmonary resistance showed a pattern of progressive worsening. Acute intravenous fasudil markedly reduced the elevated pressure and resistance at all time points tested. The percentage of severely occluded small pulmonary arteries showed a similar pattern of progression to that of right ventricular systolic pressure. These small vessels were occluded predominantly with nonplexiform-type neointimal formation except for the 13-wk time point. There was no severe occlusion in larger arteries until the 13-wk time point, when significant numbers of vessels were occluded with plexiform-type neointima. The Sugen5416/hypoxia/normoxia-exposed rat shows a pattern of chronic hemodynamic progression similar to that observed in pulmonary arterial hypertension patients. In addition to vasoconstriction, nonplexiform-type neointimal occlusion of small arteries appears to contribute significantly to the early phase of pulmonary arterial hypertension development, and plexiform-type larger vessel occlusion may play a role in the late deterioration.


Subject(s)
Hemodynamics , Hypertension, Pulmonary/physiopathology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Familial Primary Pulmonary Hypertension , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypoxia/complications , Indoles/toxicity , Male , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pyrroles/toxicity , Rats , Rats, Sprague-Dawley , Vasodilator Agents/pharmacology
5.
Am J Pathol ; 183(6): 1779-1788, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24113457

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by elevated pulmonary arterial pressure with lumen-occluding neointimal and plexiform lesions. Activation of store-operated calcium entry channels promotes contraction and proliferation of lung vascular cells. TRPC4 is a ubiquitously expressed store-operated calcium entry channel, but its role in PAH is unknown. We tested the hypothesis that TRPC4 promotes pulmonary arterial constriction and occlusive remodeling, leading to right ventricular failure in severe PAH. Severe PAH was induced in Sprague-Dawley rats and in wild-type and TRPC4-knockout Fischer 344 rats by a single subcutaneous injection of SU5416 [SU (semaxanib)], followed by hypoxia exposure (Hx; 10% O2) for 3 weeks and then a return to normoxia (Nx; 21% O2) for 3 to 10 additional weeks (SU/Hx/Nx). Although rats of both backgrounds exhibited indistinguishable pulmonary hypertensive responses to SU/Hx/Nx, Fischer 344 rats died within 6 to 8 weeks. Normoxic and hypertensive TRPC4-knockout rats recorded hemodynamic parameters similar to those of their wild-type littermates. However, TRPC4 inactivation conferred a striking survival benefit, due in part to preservation of cardiac output. Histological grading of vascular lesions revealed a reduction in the density of severely occluded small pulmonary arteries and in the number of plexiform lesions in TRPC4-knockout rats. TRPC4 inactivation therefore provides a survival benefit in severe PAH, associated with a decrease in the magnitude of occlusive remodeling.


Subject(s)
Hypertension, Pulmonary , TRPC Cation Channels , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Cardiac Output/drug effects , Disease Models, Animal , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/therapy , Indoles/adverse effects , Indoles/pharmacology , Male , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pyrroles/adverse effects , Pyrroles/pharmacology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Time Factors
6.
Mol Pharm ; 11(12): 4374-84, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25333706

ABSTRACT

This study sought to develop a liposomal delivery system of fasudil--an investigational drug for the treatment of pulmonary arterial hypertension (PAH)--that will preferentially accumulate in the PAH lungs. Liposomal fasudil was prepared by film-hydration method, and the drug was encapsulated by active loading. The liposome surface was coated with a targeting moiety, CARSKNKDC, a cyclic peptide; the liposomes were characterized for size, polydispersity index, zeta potential, and storage and nebulization stability. The in vitro drug release profiles and uptake by TGF-ß activated pulmonary arterial smooth muscle cells (PASMC) and alveolar macrophages were evaluated. The pharmacokinetics were monitored in male Sprague-Dawley rats, and the pulmonary hemodynamics were studied in acute and chronic PAH rats. The size, polydispersity index (PDI), and zeta potential of the liposomes were 206-216 nm, 0.058-0.084, and -20-42.7 mV, respectively. The formulations showed minimal changes in structural integrity when nebulized with a commercial microsprayer. The optimized formulation was stable for >4 weeks when stored at 4 °C. Fasudil was released in a continuous fashion over 120 h with a cumulative release of 76%. Peptide-linked liposomes were taken up at a higher degree by TGF-ß activated PASMCs; but alveolar macrophages could not engulf peptide-coated liposomes. The formulations did not injure the lungs; the half-life of liposomal fasudil was 34-fold higher than that of plain fasudil after intravenous administration. Peptide-linked liposomal fasudil, as opposed to plain liposomes, reduced the mean pulmonary arterial pressure by 35-40%, without influencing the mean systemic arterial pressure. This study establishes that CAR-conjugated inhalable liposomal fasudil offers favorable pharmacokinetics and produces pulmonary vasculature specific dilatation.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Hypertension, Pulmonary/drug therapy , Liposomes/chemistry , Peptides/chemistry , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/chemistry , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Animals , Male , Rats , Rats, Sprague-Dawley , Vasodilator Agents/chemistry , Vasodilator Agents/therapeutic use
7.
J Cardiovasc Pharmacol ; 64(5): 473-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25084082

ABSTRACT

The efficacy of endothelin (ET) receptor antagonist bosentan in patients with severe pulmonary arterial hypertension (PAH) remains limited, partly because its higher doses for potential blockade of ET receptors have never been tested due to liver dysfunction. We hypothesized that rigorous blockade of ET receptors using the novel dual ET receptor antagonist macitentan would be effective in treating severe PAH without major side effects in a preclinical model appropriately representing the human disorder. In normal rats, 30 mg·kg·d of macitentan completely abolished big ET-1-induced increases in right ventricle (RV) systolic pressure. Adult male rats were injected with SU5416, a vascular endothelial growth factor blocker, and exposed to hypoxia for 3 weeks and then to normoxia for an additional 5 weeks (total 8 weeks). In intrapulmonary arterial rings isolated from rats with severe PAH, macitentan concentration dependently inhibited ET-1-induced contraction. Long-term treatment with macitentan (30 mg·kg·d, from week 3 to 8) reversed the high RV systolic pressure with preserved cardiac output. Development of RV hypertrophy, luminal occlusive lesions and medial wall thickening were also significantly improved without increasing serum levels of liver enzymes by macitentan. In conclusion, efficacious blockade of ET receptors with macitentan would reverse severe PAH without major adverse effects.


Subject(s)
Endothelin A Receptor Antagonists/pharmacology , Endothelin B Receptor Antagonists/pharmacology , Hypertension, Pulmonary/drug therapy , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelin A Receptor Antagonists/administration & dosage , Endothelin A Receptor Antagonists/toxicity , Endothelin B Receptor Antagonists/administration & dosage , Endothelin B Receptor Antagonists/toxicity , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/drug therapy , Indoles/pharmacology , Male , Pyrimidines/administration & dosage , Pyrimidines/toxicity , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Sulfonamides/administration & dosage , Sulfonamides/toxicity , Time Factors , Vascular Endothelial Growth Factor A/antagonists & inhibitors
8.
Am J Physiol Heart Circ Physiol ; 304(12): H1708-18, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23585128

ABSTRACT

Current therapy of pulmonary arterial hypertension (PAH) is inadequate. Dehydroepiandrosterone (DHEA) effectively treats experimental pulmonary hypertension in chronically hypoxic and monocrotaline-injected rats. Contrary to these animal models, SU5416/hypoxia/normoxia-exposed rats develop a more severe form of occlusive pulmonary arteriopathy and right ventricular (RV) dysfunction that is indistinguishable from the human disorder. Thus, we tested the effects of DHEA treatment on PAH and RV structure and function in this model. Chronic (5 wk) DHEA treatment significantly, but moderately, reduced the severely elevated RV systolic pressure. In contrast, it restored the impaired cardiac index to normal levels, resulting in an improved cardiac function, as assessed by echocardiography. Moreover, DHEA treatment inhibited RV capillary rarefaction, apoptosis, fibrosis, and oxidative stress. The steroid decreased NADPH levels in the RV. As a result, the reduced reactive oxygen species production in the RV of these rats was reversed by NADPH supplementation. Mechanistically, DHEA reduced the expression and activity of Rho kinases in the RV, which was associated with the inhibition of cardiac remodeling-related transcription factors STAT3 and NFATc3. These results show that DHEA treatment slowed the progression of severe PAH in SU5416/hypoxia/normoxia-exposed rats and protected the RV against apoptosis and fibrosis, thus preserving its contractile function. The antioxidant activity of DHEA, by depleting NADPH, plays a central role in these cardioprotective effects.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Heart Ventricles/pathology , Hypertension, Pulmonary/drug therapy , Pulmonary Artery/pathology , Ventricular Dysfunction/drug therapy , Animals , Apoptosis , Blood Pressure/drug effects , Fibrosis , Gene Expression , Heart Ventricles/metabolism , Hypertension, Pulmonary/etiology , Hypoxia/complications , Indoles/toxicity , Male , NADP/metabolism , NFATC Transcription Factors/antagonists & inhibitors , Oxidative Stress , Pyrroles/toxicity , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/antagonists & inhibitors , Ventricular Dysfunction/etiology , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
9.
Mol Pharm ; 10(5): 1655-67, 2013 May 06.
Article in English | MEDLINE | ID: mdl-23485062

ABSTRACT

This study sought to investigate the efficacy of a noninvasive and long acting polymeric particle based formulation of prostaglandin E1 (PGE1), a potent pulmonary vasodilator, in alleviating the signs of pulmonary hypertension (PH) and reversing the biochemical changes that occur in the diseased lungs. PH rats, developed by a single subcutaneous injection of monocrotaline (MCT), were treated with two types of polymeric particles of PGE1, porous and nonporous, and intratracheal or intravenous plain PGE1. For chronic studies, rats received either intratracheal porous poly(lactic-co-glycolic acid) (PLGA) particles, once- or thrice-a-day, or plain PGE1 thrice-a-day for 10 days administered intratracheally or intravenously. The influence of formulations on disease progression was studied by measuring the mean pulmonary arterial pressure (MPAP), evaluating right ventricular hypertrophy and assessing various molecular and cellular makers including the degree of muscularization, platelet aggregation, matrix metalloproteinase-2 (MMP-2), and proliferating cell nuclear antigen (PCNA). Both plain PGE1 and large porous particles of PGE1 reduced MPAP and right ventricular hypertrophy (RVH) in rats that received the treatments for 10 days. Polymeric porous particles of PGE1 produced the same effects at a reduced dosing frequency compared to plain PGE1 and caused minimal off-target effects on systemic hemodynamics. Microscopic and immunohistochemical studies revealed that porous particles of PGE1 also reduced the degree of muscularization, von Willebrand factor (vWF), and PCNA expression in the lungs of PH rats. Overall, our study suggests that PGE1 loaded inhalable particulate formulations improve PH symptoms and arrest the progression of disease at a reduced dosing frequency compared to plain PGE1.


Subject(s)
Alprostadil/administration & dosage , Hypertension, Pulmonary/drug therapy , Administration, Inhalation , Animals , Delayed-Action Preparations , Disease Models, Animal , Disease Progression , Drug Carriers/chemistry , Hemodynamics/drug effects , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/pathology , Lactic Acid/chemistry , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Vasodilator Agents/administration & dosage , von Willebrand Factor/metabolism
10.
Am J Pathol ; 178(6): 2489-95, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21549345

ABSTRACT

Pulmonary arterial hypertension (PAH) is a disorder of the pulmonary vasculature associated with elevated pulmonary vascular resistance. Despite recent advances in the treatment of PAH, with eight approved clinical therapies and additional therapies undergoing clinical trials, PAH remains a serious life-threatening condition. The lack of pulmonary vascular selectivity and associated systemic adverse effects of these therapies remain the main obstacles to successful treatment. Peptide-mediated drug delivery that specifically targets the vasculature of PAH lungs may offer a solution to the lack of drug selectivity. Herein, we show highly selective targeting of rat PAH lesions by a novel cyclic peptide, CARSKNKDC (CAR). Intravenous administration of CAR peptide resulted in intense accumulation of the peptide in monocrotaline-induced and SU5416/hypoxia-induced hypertensive lungs but not in healthy lungs or other organs of PAH rats. CAR homed to all layers of remodeled pulmonary arteries, ie, endothelium, neointima, medial smooth muscle, and adventitia, in the hypertensive lungs. CAR also homed to capillary vessels and accumulated in the interstitial space of the PAH lungs, manifesting its extravasation activity. These results demonstrated the remarkable ability of CAR to selectively target PAH lung vasculature and effectively penetrate and spread throughout the diseased lung tissue. These results suggest the clinical utility of CAR in the targeted delivery of therapeutic compounds and imaging probes to PAH lungs.


Subject(s)
Drug Delivery Systems , Hypertension, Pulmonary/pathology , Peptides/pharmacology , Pulmonary Artery/pathology , Amino Acid Sequence , Animals , Humans , Hypertension, Pulmonary/complications , Hypoxia/complications , Indoles/pharmacology , Lung/blood supply , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Molecular Sequence Data , Monocrotaline , Peptides/administration & dosage , Peptides/chemistry , Pulmonary Artery/drug effects , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Time Factors
11.
Am J Respir Cell Mol Biol ; 45(4): 804-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21378262

ABSTRACT

Tyrosine kinase inhibitors are promising for the treatment of severe pulmonary hypertension. Their therapeutic effects are postulated to be due to inhibition of cell growth-related kinases and attenuation of vascular remodeling. Their potential vasodilatory activities have not been explored. Vasorelaxant effects of the tyrosine kinase inhibitors imatinib, sorafenib, and nilotinib were examined in isolated pulmonary arterial rings from normal and pulmonary hypertensive rats. Phosphorylation of myosin light chain phosphatase and myosin light chain was assessed by Western blots. Acute hemodynamic effects of imatinib were tested in the pulmonary hypertensive rats. In normal pulmonary arteries, imatinib reversed serotonin- and U46619-induced contractions in a concentration-dependent and endothelium-independent manner. Sorafenib and nilotinib relaxed U46619-induced contraction. Imatinib inhibited activation of myosin phosphatase induced by U46619 in normal pulmonary arteries. All three tyrosine kinase inhibitors concentration-dependently and completely reversed the spontaneous contraction of hypertensive pulmonary arterial rings unmasked by inhibition of nitric oxide synthase. Acute intravenous administration of imatinib reduced high right ventricular systolic pressure in pulmonary hypertensive rats, with little effect on left ventricular systolic pressure and cardiac output. We conclude that tyrosine kinase inhibitors have potent pulmonary vasodilatory activity, which could contribute to their long-term beneficial effect against pulmonary hypertension. Vascular smooth muscle relaxation mediated via activation of myosin light chain phosphatase (Ca(2+) desensitization) appears to play a role in the imatinib-induced pulmonary vasodilation.


Subject(s)
Antihypertensive Agents/pharmacology , Hypertension, Pulmonary/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pulmonary Artery/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Benzamides , Benzenesulfonates/pharmacology , Blotting, Western , Calcium/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/physiopathology , Imatinib Mesylate , Male , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds , Phosphorylation , Piperazines/pharmacology , Protein-Tyrosine Kinases/metabolism , Pulmonary Artery/physiopathology , Pyridines/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Sorafenib , Ventricular Function, Left/drug effects , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects
12.
Circulation ; 121(25): 2747-54, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20547927

ABSTRACT

BACKGROUND: The plexiform lesion is the hallmark of severe pulmonary arterial hypertension. However, its genesis and hemodynamic effects are largely unknown because of the limited availability of lung tissue samples from patients with pulmonary arterial hypertension and the lack of appropriate animal models. This study investigated whether rats with severe progressive pulmonary hypertension developed plexiform lesions. METHODS AND RESULTS: After a single subcutaneous injection of the vascular endothelial growth factor receptor blocker Sugen 5416, rats were exposed to hypoxia for 3 weeks. They were then returned to normoxia for an additional 10 to 11 weeks. Hemodynamic and histological examinations were performed at 13 to 14 weeks after the Sugen 5416 injection. All rats developed pulmonary hypertension (right ventricular systolic pressure approximately 100 mm Hg) and severe pulmonary arteriopathy, including concentric neointimal and complex plexiform-like lesions. There were 2 patterns of complex lesion formation: a lesion forming within the vessel lumen (stalk-like) and another that projected outside the vessel (aneurysm-like). Immunohistochemical analyses showed that these structures had cellular and molecular features closely resembling human plexiform lesions. CONCLUSIONS: Severe, sustained pulmonary hypertension in a very late stage of the Sugen 5416/hypoxia/normoxia-exposed rat is accompanied by the formation of lesions that are indistinguishable from the pulmonary arteriopathy of human pulmonary arterial hypertension. This unique model provides a new and rigorous approach for investigating the genesis, hemodynamic effects, and reversibility of plexiform and other occlusive lesions in pulmonary arterial hypertension.


Subject(s)
Disease Models, Animal , Hypertension, Pulmonary/pathology , Animals , Arteries/pathology , Hypertension, Pulmonary/etiology , Hypoxia , Lung/blood supply , Rats , Vascular Endothelial Growth Factor A/antagonists & inhibitors
13.
BMC Immunol ; 12: 67, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22171643

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) refers to a spectrum of diseases with elevated pulmonary artery pressure. Pulmonary arterial hypertension (PAH) is a disease category that clinically presents with severe PH and that is histopathologically characterized by the occlusion of pulmonary arterioles, medial muscular hypertrophy, and/or intimal fibrosis. PAH occurs with a secondary as well as a primary onset. Secondary PAH is known to be complicated with immunological disorders. The aim of the present study is to histopathologically and genetically characterize a new animal model of PAH and clarify the role of OX40 ligand in the pathogenesis of PAH. RESULTS: Spontaneous onset of PAH was stably identified in mice with immune abnormality because of overexpression of the tumor necrosis factor (TNF) family molecule OX40 ligand (OX40L). Histopathological and physical examinations revealed the onset of PAH-like disorders in the C57BL/6 (B6) strain of OX40L transgenic mice (B6.TgL). Comparative analysis performed using different strains of transgenic mice showed that this onset depends on the presence of OX40L in the B6 genetic background. Genetic analyses demonstrated a susceptibility locus of a B6 allele to this onset on chromosome 5. Immunological analyses revealed that the excessive OX40 signals in TgL mice attenuates expansion of regulatory T cells the B6 genetic background, suggesting an impact of the B6 genetic background on the differentiation of regulatory T cells. CONCLUSION: Present findings suggest a role for the OX40L-derived immune response and epistatic genetic effect in immune-mediated pathogenesis of PAH.


Subject(s)
Epistasis, Genetic , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/immunology , OX40 Ligand/genetics , Signal Transduction , Animals , Blood Pressure , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Female , Gene Expression , Genetic Predisposition to Disease , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/pathology , Immunologic Memory , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , OX40 Ligand/metabolism , Phenotype , Quantitative Trait Loci
14.
Am J Respir Crit Care Med ; 182(12): 1554-62, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20833821

ABSTRACT

The Division of Lung Diseases of the National Heart, Lung, and Blood Institute, with the Office of Rare Diseases Research, held a workshop to identify priority areas and strategic goals to enhance and accelerate research that will result in improved understanding of the lung vasculature, translational research needs, and ultimately the care of patients with pulmonary vascular diseases. Multidisciplinary experts with diverse experience in laboratory, translational, and clinical studies identified seven priority areas and discussed limitations in our current knowledge, technologies, and approaches. The focus for future research efforts include the following: (1) better characterizing vascular genotype-phenotype relationships and incorporating systems biology approaches when appropriate; (2) advancing our understanding of pulmonary vascular metabolic regulatory signaling in health and disease; (3) expanding our knowledge of the biologic relationships between the lung circulation and circulating elements, systemic vascular function, and right heart function and disease; (4) improving translational research for identifying disease-modifying therapies for the pulmonary hypertensive diseases; (5) establishing an appropriate and effective platform for advancing translational findings into clinical studies testing; and (6) developing the specific technologies and tools that will be enabling for these goals, such as question-guided imaging techniques and lung vascular investigator training programs. Recommendations from this workshop will be used within the Lung Vascular Biology and Disease Extramural Research Program for planning and strategic implementation purposes.


Subject(s)
Biomedical Research/methods , Guidelines as Topic , Lung Diseases/physiopathology , Lung/blood supply , Pulmonary Circulation , Humans
15.
Am J Pathol ; 174(3): 782-96, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19234135

ABSTRACT

Neprilysin is a transmembrane metalloendopeptidase that degrades neuropeptides that are important for both growth and contraction. In addition to promoting carcinogenesis, decreased levels of neprilysin increases inflammation and neuroendocrine cell hyperplasia, which may predispose to vascular remodeling. Early pharmacological studies showed a decrease in chronic hypoxic pulmonary hypertension with neprilysin inhibition. We used a genetic approach to test the alternate hypothesis that neprilysin depletion increases chronic hypoxic pulmonary hypertension. Loss of neprilysin had no effect on baseline airway or alveolar wall architecture, vessel density, cardiac function, hematocrit, or other relevant peptidases. Only lung neuroendocrine cell hyperplasia and a subtle neuropeptide imbalance were found. After chronic hypoxia, neprilysin-null mice exhibited exaggerated pulmonary hypertension and striking increases in muscularization of distal vessels. Subtle thickening of proximal media/adventitia not typically seen in mice was also detected. In contrast, adaptive right ventricular hypertrophy was less than anticipated. Hypoxic wild-type pulmonary vessels displayed close temporal and spatial relationships between decreased neprilysin and increased cell growth. Smooth muscle cells from neprilysin-null pulmonary arteries had increased proliferation compared with controls, which was decreased by neprilysin replacement. These data suggest that neprilysin may be protective against chronic hypoxic pulmonary hypertension in the lung, at least in part by attenuating the growth of smooth muscle cells. Lung-targeted strategies to increase neprilysin levels could have therapeutic benefits in the treatment of this disorder.


Subject(s)
Hypertension, Pulmonary/pathology , Hypoxia/genetics , Mice, Knockout , Neprilysin/deficiency , Pulmonary Artery/pathology , Pulmonary Circulation/physiology , Animals , Cell Division , Chronic Disease , DNA Primers , Genetic Predisposition to Disease , Genotype , Hemodynamics , Hypertension, Pulmonary/genetics , Hypoxia/pathology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Neprilysin/genetics
16.
Adv Exp Med Biol ; 661: 299-308, 2010.
Article in English | MEDLINE | ID: mdl-20204738

ABSTRACT

Rho kinase-mediated vasoconstriction rather than fixed arterial wall thickening is responsible for increased pulmonary vascular resistance and pulmonary hypertension in chronically hypoxic and monocrotaline-injected rats. In the absence of vascular tone, the medial and adventitial thickening in these models has only minimal impact on the cross-sectional area of the pulmonary arterial bed. In contrast, increased pulmonary vascular resistance in left-pneumonectomized plus monocrotaline-injected rats and VEGF receptor blocker-injected plus chronic hypoxia rats is attributable to both Rho kinase-mediated vasoconstriction and formation of lumen obliterating lesions in small pulmonary arteries. The upstream signals responsible for activation of RhoA/Rho kinase signaling in hypertensive pulmonary arteries and whether or not they differ in different forms of pulmonary hypertension are unclear. The RhoA/Rho kinase pathway is a convergence point of several different vasoconstrictor signals, including those mediated by G protein-coupled receptors, receptor tyrosine kinases, and integrin clustering. Both isoforms of Rho kinase can also be constitutively activated by cleavage, and cleaved Rho kinase 1 has been detected in the hypertensive lungs of left-pneumonectomized plus monocrotaline-injected rats. That such diverse stimuli can lead to activation of Rho kinase, which may cause hypercontraction of smooth muscle by promoting both actomyosin interaction and remodeling of the cytoskeleton, may explain why in various rat models of pulmonary hypertension Rho kinase inhibitors are more effective pulmonary vasodilators than conventional agents such as nitric oxide, prostacyclin, and nifedipine. We suspect the same will be true in at least some forms of human pulmonary arterial hypertension.


Subject(s)
Hypertension, Pulmonary/physiopathology , Vasoconstriction/physiology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Humans , Hypertension, Pulmonary/pathology , Lung/blood supply , Lung/metabolism , Lung/pathology , Muscle Contraction/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Signal Transduction/physiology
17.
Circ Res ; 100(6): 923-9, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17332430

ABSTRACT

Vascular remodeling, rather than vasoconstriction, is believed to account for high vascular resistance in severe pulmonary arterial hypertension (PAH). We have found previously that acute Rho kinase inhibition nearly normalizes PAH in chronically hypoxic rats that have no occlusive neointimal lesions. Here we examined whether Rho kinase-mediated vasoconstriction was also important in a rat model of severe occlusive PAH. Adult rats were exposed to chronic hypoxia ( approximately 10% O(2)) after subcutaneous injection of the vascular endothelial growth factor receptor inhibitor SUGEN 5416. Hemodynamic measurements were made in anesthetized rats after 2 weeks of hypoxia (early group) and 3 weeks of hypoxia plus 2 weeks of normoxia (late group). Both groups developed PAH, with greater severity in the late group. In the early group, intravenous fasudil was more effective than intravenous bradykinin, inhaled NO, or intravenous iloprost in reducing right ventricular systolic pressure. Despite more occlusive vascular lesions, fasudil also markedly reduced right ventricular systolic pressure in late-stage rats. Blood-perfused lungs from late-stage rats showed spontaneous vasoconstriction, which was reversed partially by the endothelin A receptor blocker BQ123 and completely by fasudil or Y-27632. Phosphorylation of MYPT1, a downstream target of Rho kinase, was increased in lungs from both groups of rats, and fasudil (intravenous) reversed the increased phosphorylation in the late group. Thus, in addition to structural occlusion, Rho kinase-mediated vasoconstriction is an important component of severe PAH in SUGEN 5416/hypoxia-exposed rats, and PAH can be significantly reduced in the setting of a severely remodeled lung circulation if an unconventional vasodilator is used.


Subject(s)
Hypertension, Pulmonary/physiopathology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Pulmonary Artery/physiopathology , Vasoconstriction , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Disease Models, Animal , Disease Progression , Endothelin A Receptor Antagonists , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/enzymology , Hypoxia , Indoles , Male , Myosin-Light-Chain Phosphatase/metabolism , Organ Culture Techniques , Peptides, Cyclic/pharmacology , Phosphorylation/drug effects , Pulmonary Artery/enzymology , Pyrroles , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vasoconstriction/drug effects , Vasodilator Agents/pharmacology , rho-Associated Kinases
18.
Cardiovasc Res ; 115(3): 658-668, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30239623

ABSTRACT

AIMS: Recent accumulating evidence suggests that sterile inflammation plays a crucial role in the progression of various cardiovascular diseases. However, its contribution to right ventricular (RV) dysfunction remains unknown. The aim of this study was to elucidate whether toll-like receptor 9 (TLR9)-NF-κB-mediated sterile inflammation plays a critical role in the pathogenesis of RV dysfunction. METHODS AND RESULTS: We performed main pulmonary artery banding (PAB) in rats to induce RV pressure overload and dysfunction. On Day 14 after PAB, the pressure overload impaired RV function as indicated by increased RV end-diastolic pressure concomitant with macrophage infiltration and fibrosis, as well as maximal activation of NF-κB and TLR9. Short-term administration (days 14-16 after PAB) of a specific TLR9 inhibitor, E6446, or an NF-κB inhibitor, pyrrolidine dithiocarbamate (PDTC) significantly attenuated NF-κB activation. Furthermore, long-term administration of E6446 (treatment: days 14-28) or PDTC (prevention: days -1 to 28; treatment: days 14 to 28) improved RV dysfunction associated with mitigated macrophage infiltration and fibrosis in right ventricle and decreased serum brain natriuretic peptide levels. CONCLUSION: Inhibition of TLR9-NF-κB pathway-mediated sterile inflammation improved PAB-induced RV dysfunction in rats. This pathway plays a major role in the progression of pressure overload-induced RV dysfunction and is potentially a novel therapeutic target for the disorder.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation Mediators/antagonists & inhibitors , Inflammation/metabolism , NF-kappa B/antagonists & inhibitors , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Toll-Like Receptor 9/antagonists & inhibitors , Ventricular Dysfunction, Right/drug therapy , Ventricular Function, Right , Ventricular Pressure , Animals , Fibrosis , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , NF-kappa B/metabolism , Rats, Sprague-Dawley , Signal Transduction , Toll-Like Receptor 9/metabolism , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology , Ventricular Remodeling/drug effects
19.
Methods Enzymol ; 439: 191-204, 2008.
Article in English | MEDLINE | ID: mdl-18374166

ABSTRACT

There is current controversy regarding whether vasoconstriction plays a significant role in the elevated pressure of severe, advanced stages of pulmonary hypertension. Results of acute vasodilator testing using conventional vasodilators in such patients suggest there is only a minor contribution of vasoconstriction. However, there is a possibility that these results may underestimate the contribution of vasoconstriction because the most effective vasodilators have not yet been tested. This issue has not been addressed even experimentally, due mainly to a lack of appropriate animal models. A few animal models that mimic the pathology of human severe pulmonary hypertension more closely (i.e., development of occlusive neointimal lesions in small pulmonary arteries/arterioles) have been introduced, including rat models of left lung pneumonectomy plus monocrotaline injection and vascular endothelial growth factor inhibition plus exposure to chronic hypoxia. We have observed that Rho kinase inhibitors, a novel class of potent vasodilators, reduce the high pulmonary artery pressure of these models acutely and markedly, suggesting that vasoconstriction can significantly be involved in pulmonary hypertension with severely remodeled (occluded) pulmonary vessels. This chapter describes methods used for evaluation of the involvement of Rho kinase-mediated vasoconstriction in rat models of pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary/physiopathology , Vasoconstriction/physiology , rho-Associated Kinases/physiology , Animals , Bacterial Toxins/pharmacology , Disease Models, Animal , Hemolysin Proteins/pharmacology , Lung/metabolism , Protein Phosphatase 1/metabolism , Protein Transport , Pulmonary Artery/drug effects , Rats , Signal Transduction/physiology , rho-Associated Kinases/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
20.
Cardiovasc Res ; 74(3): 377-87, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17346686

ABSTRACT

OBJECTIVE: It has been reported that dehydroepiandrosterone is a pulmonary vasodilator and inhibits chronic hypoxia-induced pulmonary hypertension. Additionally, dehydroepiandrosterone has been shown to improve systemic vascular endothelial function. Thus, we hypothesized that chronic treatment with dehydroepiandrosterone would attenuate hypoxic pulmonary hypertension by enhancing pulmonary artery endothelial function. METHODS AND RESULTS: Rats were randomly assigned to five groups. Three groups received food containing 0, 0.3, or 1% dehydroepiandrosterone during a 3-wk-exposure to simulated high altitude (HA). The other 2 groups were kept at Denver's low altitude (LA) and received food containing 0 or 1% dehydroepiandrosterone. Dehydroepiandrosterone dose-dependently inhibited hypoxic pulmonary hypertension (mean pulmonary artery pressures after treatment with 0, 0.3, and 1% dehydroepiandrosterone=45+/-5, 33+/-2*, and 25+/-1*# mmHg, respectively. *P<0.05 vs. 0% and # vs. 0.3%). Dehydroepiandrosterone (1%, 3 wks) treatment started after rats had been exposed to 3-wk hypoxia also effectively reversed established hypoxic pulmonary hypertension. Pulmonary artery rings isolated from both LA and HA rats treated with 1% dehydroepiandrosterone showed enhanced relaxations to acetylcholine and sodium nitroprusside, but not to 8-bromo-cGMP. In the pulmonary artery tissue from dehydroepiandrosterone-treated LA and HA rats, soluble guanylate cyclase, but not endothelial nitric oxide synthase, protein levels were increased. CONCLUSION: These results indicate that the protective effect of dehydroepiandrosterone against hypoxic pulmonary hypertension may involve upregulation of pulmonary artery soluble guanylate cyclase protein expression and augmented pulmonary artery vasodilator responsiveness to nitric oxide.


Subject(s)
Dehydroepiandrosterone/therapeutic use , Guanylate Cyclase/metabolism , Hypertension, Pulmonary/drug therapy , Pulmonary Artery/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Up-Regulation , Acetylcholine/pharmacology , Animals , Blotting, Western , Cyclic GMP/pharmacology , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone Sulfate/blood , Dehydroepiandrosterone Sulfate/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Estradiol/blood , Guanylate Cyclase/analysis , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , In Vitro Techniques , Lung/enzymology , Male , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/analysis , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitroprusside/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/analysis , Soluble Guanylyl Cyclase , Testosterone/blood , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL