Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Immunol ; 197(2): 470-9, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27279372

ABSTRACT

Pulmonary alveolar proteinosis (PAP) is a rare lung syndrome caused by the accumulation of surfactants in the alveoli. The most prevalent clinical form of PAP is autoimmune PAP (aPAP) whereby IgG autoantibodies neutralize GM-CSF. GM-CSF is a pleiotropic cytokine that promotes the differentiation, survival, and activation of alveolar macrophages, the cells responsible for surfactant degradation. IgG-mediated neutralization of GM-CSF thereby inhibits alveolar macrophage homeostasis and function, leading to surfactant accumulation and innate immunodeficiency. Importantly, there are no rodent models for this disease; therefore, underlying immune mechanisms regulating GM-CSF-specific IgG in aPAP are not well understood. In this article, we identify that autoimmune-prone Rasgrp1-deficient mice develop aPAP: 1) Rasgrp1-deficient mice exhibit reduced pulmonary compliance and lung histopathology characteristic of PAP; 2) alveolar macrophages from Rasgrp1-deficient mice are enlarged and exhibit reduced surfactant degradation; 3) the concentration of GM-CSF-specific IgG is elevated in both serum and bronchoalveolar lavage fluid from Rasgrp1-deficient mice; 4) GM-CSF-specific IgG is capable of neutralizing GM-CSF bioactivity; and 5) Rasgrp1-deficient mice also lacking CD275/ICOSL, a molecule necessary for conventional T cell-dependent Ab production, have reduced GM-CSF-specific autoantibody and do not develop PAP. Collectively, these studies reveal that Rasgrp1-deficient mice, to our knowledge, represent the first rodent model for aPAP.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/immunology , Disease Models, Animal , Guanine Nucleotide Exchange Factors/deficiency , Pulmonary Alveolar Proteinosis/immunology , Animals , Autoantigens/immunology , Autoimmune Diseases/genetics , Enzyme-Linked Immunosorbent Assay , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Guanine Nucleotide Exchange Factors/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveolar Proteinosis/genetics , Real-Time Polymerase Chain Reaction
2.
Am J Physiol Lung Cell Mol Physiol ; 308(3): L287-300, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25480333

ABSTRACT

Severe pulmonary hypertension is a debilitating disease with an alarmingly low 5-yr life expectancy. Hypoxia, one of the causes of pulmonary hypertension, elicits constriction and remodeling of the pulmonary arteries. We now know that pulmonary arterial remodeling is a consequence of hyperplasia and hypertrophy of pulmonary artery smooth muscle (PASM), endothelial, myofibroblast, and stem cells. However, our knowledge about the mechanisms that cause these cells to proliferate and hypertrophy in response to hypoxic stimuli is still incomplete, and, hence, the treatment for severe pulmonary arterial hypertension is inadequate. Here we demonstrate that the activity and expression of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the pentose phosphate pathway, are increased in hypoxic PASM cells and in lungs of chronic hypoxic rats. G6PD overexpression and -activation is stimulated by H2O2. Increased G6PD activity contributes to PASM cell proliferation by increasing Sp1 and hypoxia-inducible factor 1α (HIF-1α), which directs the cells to synthesize less contractile (myocardin and SM22α) and more proliferative (cyclin A and phospho-histone H3) proteins. G6PD inhibition with dehydroepiandrosterone increased myocardin expression in remodeled pulmonary arteries of moderate and severe pulmonary hypertensive rats. These observations suggest that altered glucose metabolism and G6PD overactivation play a key role in switching the PASM cells from the contractile to synthetic phenotype by increasing Sp1 and HIF-1α, which suppresses myocardin, a key cofactor that maintains smooth muscle cell in contractile state, and increasing hypoxia-induced PASM cell growth, and hence contribute to pulmonary arterial remodeling and pathogenesis of pulmonary hypertension.


Subject(s)
Glucosephosphate Dehydrogenase/genetics , Hypertension, Pulmonary/enzymology , Myocytes, Smooth Muscle/enzymology , Pulmonary Artery/pathology , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Hypoxia , Cell Proliferation , Enzyme Induction , Gene Expression , Glucosephosphate Dehydrogenase/metabolism , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nuclear Proteins/metabolism , Protein Biosynthesis , Rats , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Trans-Activators/metabolism , Up-Regulation
3.
Am J Physiol Lung Cell Mol Physiol ; 307(7): L545-56, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25063801

ABSTRACT

Although hypoxia is detrimental to most cell types, it aids survival of progenitor cells and is associated with diseases like cancer and pulmonary hypertension in humans. Therefore, understanding the underlying mechanisms that promote survival of progenitor cells in hypoxia and then developing novel therapies to stop their growth in hypoxia-associated human diseases is important. Here we demonstrate that the proliferation and growth of human CD133(+) progenitor cells, which contribute to tumorigenesis and the development of pulmonary hypertension, are increased when cultured under hypoxic conditions. Furthermore, glucose-6-phosphate dehydrogenase (G6PD) activity was increased threefold in hypoxic CD133(+) cells. The increased G6PD activity was required for CD133(+) cell proliferation, and their growth was arrested by G6PD inhibition or knockdown. G6PD activity upregulated expression of HIF1α, cyclin A, and phospho-histone H3, thereby promoting CD133(+) cell dedifferentiation and self-renewal and altering cell cycle regulation. When CD133(+) cells were cocultured across a porous membrane from pulmonary artery smooth muscle cells (PASMCs), G6PD-dependent H2O2 production and release by PASMCs recruited CD133(+) cells to the membrane, where they attached and expressed smooth muscle markers (α-actin and SM22α). Inhibition of G6PD reduced smooth muscle marker expression in CD133(+) cells under normoxia but not hypoxia. In vivo, CD133(+) cells colocalized with G6PD(+) cells in the perivascular region of lungs from rats with hypoxia-induced pulmonary hypertension. Finally, inhibition of G6PD by dehydroepiandrosterone in pulmonary arterial hypertensive rats nearly abolished CD133(+) cell accumulation around pulmonary arteries and the formation of occlusive lesions. These observations suggest G6PD plays a key role in increasing hypoxia-induced CD133(+) cell survival in hypertensive lungs that differentiate to smooth muscle cells and contribute to pulmonary arterial remodeling during development of pulmonary hypertension.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation , Glucosephosphate Dehydrogenase/physiology , Glycoproteins/metabolism , Hypertension, Pulmonary/enzymology , Peptides/metabolism , Stem Cells/enzymology , AC133 Antigen , Administration, Oral , Animals , Cell Differentiation , Cell Hypoxia , Coculture Techniques , Dehydroepiandrosterone/administration & dosage , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/pathology , Male , Protein Transport , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction , Stem Cells/physiology , Transforming Growth Factor beta/metabolism
4.
FASEB J ; 28(9): 4088-99, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24903275

ABSTRACT

Inadequate cell proliferation is considered a major causative factor for impaired coronary collateral growth (CCG). Proangiogenic growth factors (GFs) stimulate cell proliferation, but their administration does not promote CCG in patients. These GFs are increased in patients with metabolic syndrome and in animal models, where CCG is impaired. Here, we investigated whether excessive cell proliferation underlies impaired CCG in metabolic syndrome. Normal [Sprague-Dawley (SD)] and metabolic syndrome [James C. Russell (JCR)] rats underwent repetitive ischemia (RI; transient, repetitive coronary artery occlusion and myocardial ischemia). We have shown that CCG was maximal at d 9 of RI in SD rats but did not occur in JCR rats. The increase in cell proliferation (PCNA, Ki-67, cyclin A, phospho- cdc2, p21Waf, p27Kip) was transient (∼4-fold, d 3 RI) in SD rats but greater and sustained in JCR rats (∼8- to 6-fold, d 3-9 RI). In JCR rats, this was associated with increased and sustained miR-21 expression and accumulation of proliferating synthetic vascular smooth muscle cells in the lumen of small arterioles, which failed to undergo outward expansion. Administration of anti-miR-21 blocked RI-induced cell proliferation and significantly improved CCG in JCR rats (∼60%). miR-21-dependent excessive cell proliferation in the later stages of collateral remodeling correlates with impaired CCG in metabolic syndrome.


Subject(s)
Cell Proliferation/genetics , Collateral Circulation/physiology , Coronary Artery Disease/prevention & control , Coronary Circulation/physiology , Metabolic Syndrome/physiopathology , MicroRNAs/metabolism , Muscle, Smooth, Vascular/cytology , Myocardial Ischemia/prevention & control , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Disease Models, Animal , Immunoenzyme Techniques , Male , MicroRNAs/genetics , Muscle, Smooth, Vascular/metabolism , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Neovascularization, Pathologic/prevention & control , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
5.
Neonatology ; 103(2): 91-7, 2013.
Article in English | MEDLINE | ID: mdl-23154780

ABSTRACT

BACKGROUND: Numerous studies in cultured cells indicate that damage to mitochondrial DNA (mtDNA) dictates cellular responses to oxidant stress, yet the consequences of mtDNA damage have not been studied directly in the preterm lung. OBJECTIVE: We sought to determine whether hyperoxia-induced fetal lung dysmorphogenesis is linked to mtDNA damage and establish mtDNA repair as a potential therapeutic approach for treating lung dysplasia in the preterm neonate. METHODS: Hyperoxia-induced mtDNA damage was assessed by quantitative alkaline gel electrophoresis in normoxic (3% O2) and hyperoxic (21% O2) fetal rat lung explants. A fusion protein construct targeting the DNA repair enzyme endonuclease III (Endo III) to the mitochondria was used to augment mtDNA repair. Fetal lung branching and surfactant protein C (SFPTC) were assessed in these tissues. RESULTS: Hyperoxia induced mtDNA damage in lung explants and was accompanied by impaired branching morphogenesis and decreased SFPTC mRNA expression. Treatment of lung explants with Endo III fusion protein prevented hyperoxia-induced mtDNA damage and restored normal branching morphogenesis and SFPTC mRNA expression. CONCLUSION: These findings support the concept that mtDNA governs cellular responses to oxidant stress in the fetal lung and suggest that modulation of mtDNA repair is a potential pharmacologic strategy in the prevention of hyperoxic lung injury.


Subject(s)
DNA Damage/physiology , DNA, Mitochondrial/physiology , Hyperoxia/complications , Lung Diseases/embryology , Lung Diseases/etiology , Lung/embryology , Animals , DNA Repair/drug effects , DNA, Mitochondrial/drug effects , Deoxyribonuclease (Pyrimidine Dimer) , Endodeoxyribonucleases/administration & dosage , Endodeoxyribonucleases/genetics , Lung/ultrastructure , Morphogenesis/drug effects , Oxygen/administration & dosage , Rats , Recombinant Fusion Proteins/administration & dosage , Tissue Culture Techniques
6.
Am J Physiol Lung Cell Mol Physiol ; 303(9): L767-77, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22923644

ABSTRACT

Myoendothelial gap junctional signaling mediates pulmonary arterial endothelial cell (PAEC)-induced activation of latent TGF-ß and differentiation of cocultured pulmonary arterial smooth muscle cells (PASMCs), but the nature of the signal passing from PAECs to PASMCs through the gap junctions is unknown. Because PAECs but not PASMCs synthesize serotonin, and serotonin can pass through gap junctions, we hypothesized that the monoamine is the intercellular signal. We aimed to determine whether PAEC-derived serotonin mediates PAEC-induced myoendothelial gap junction-dependent activation of TGF-ß signaling and differentiation of PASMCs. Rat PAECs and PASMCs were monocultured or cocultured with (touch) or without (no-touch) direct cell-cell contact. In all cases, tryptophan hydroxylase 1 (Tph1) transcripts were expressed predominantly in PAECs. Serotonin was detected by immunostaining in both PAECs and PASMCs in PAEC/PASMC touch coculture but was not found in PASMCs in either PAEC/PASMC no-touch coculture or in PASMC/PASMC touch coculture. Furthermore, inhibition of gap junctions but not of the serotonin transporter in PAEC/PASMC touch coculture prevented serotonin transfer from PAECs to PASMCs. Inhibition of serotonin synthesis pharmacologically or by small interfering RNAs to Tph1 in PAECs inhibited the PAEC-induced activation of TGF-ß signaling and differentiation of PASMCs. We concluded that serotonin synthesized by PAECs is transferred through myoendothelial gap junctions into PASMCs, where it activates TGF-ß signaling and induces a more differentiated phenotype. This finding suggests a novel role of gap junction-mediated intercellular serotonin signaling in regulation of PASMC phenotype.


Subject(s)
Gap Junctions/metabolism , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/cytology , Serotonin/metabolism , Signal Transduction , Animals , Carbenoxolone/pharmacology , Cells, Cultured , Coculture Techniques , Connexin 43/genetics , Connexin 43/metabolism , Endothelial Cells/metabolism , Fenclonine/pharmacology , Gap Junctions/drug effects , Gene Expression , Male , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Organ Specificity , Pulmonary Artery/metabolism , Rats , Rats, Sprague-Dawley , Tryptophan Hydroxylase/antagonists & inhibitors , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
7.
PLoS One ; 7(6): e39009, 2012.
Article in English | MEDLINE | ID: mdl-22720015

ABSTRACT

BACKGROUND: Adult mammalian cardiac myocytes are generally assumed to be terminally differentiated; nonetheless, a small fraction of cardiac myocytes have been shown to replicate during ventricular remodeling. However, the expression of Replication Factor C (RFC; RFC140/40/38/37/36) and DNA polymerase δ (Pol δ) proteins, which are required for DNA synthesis and cell proliferation, in the adult normal and hypertrophied hearts has been rarely studied. METHODS: We performed qRT-PCR and Western blot analysis to determine the levels of RFC and Pol δ message and proteins in the adult normal cardiac myocytes and cardiac fibroblasts, as well as in adult normal and pulmonary arterial hypertension induced right ventricular hypertrophied hearts. Immunohistochemical analyses were performed to determine the localization of the re-expressed DNA replication and cell cycle proteins in adult normal (control) and hypertrophied right ventricle. We determined right ventricular cardiac myocyte polyploidy and chromosomal missegregation/aneuploidy using Fluorescent in situ hybridization (FISH) for rat chromosome 12. RESULTS: RFC40-mRNA and protein was undetectable, whereas Pol δ message was detectable in the cardiac myocytes isolated from control adult hearts. Although RFC40 and Pol δ message and protein significantly increased in hypertrophied hearts as compared to the control hearts; however, this increase was marginal as compared to the fetal hearts. Immunohistochemical analyses revealed that in addition to RFC40, proliferative and mitotic markers such as cyclin A, phospho-Aurora A/B/C kinase and phospho-histone 3 were also re-expressed/up-regulated simultaneously in the cardiac myocytes. Interestingly, FISH analyses demonstrated cardiac myocytes polyploidy and chromosomal missegregation/aneuploidy in these hearts. Knock-down of endogenous RFC40 caused chromosomal missegregation/aneuploidy and decrease in the rat neonatal cardiac myocyte numbers. CONCLUSION: Our novel findings suggest that transcription of RFC40 is suppressed in the normal adult cardiac myocytes and its insufficient re-expression may be responsible for causing chromosomal missegregation/aneuploidy and in cardiac myocytes during right ventricular hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Chromosomes , Down-Regulation , Myocardium/metabolism , Replication Protein C/metabolism , Animals , Animals, Newborn , Cardiomegaly/pathology , Cells, Cultured , DNA Polymerase III/metabolism , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Myocardium/cytology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction
8.
Am J Physiol Lung Cell Mol Physiol ; 301(4): L527-35, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21803868

ABSTRACT

Myoendothelial gap junctions are involved in regulating systemic arterial smooth muscle cell phenotype and function, but their role in the regulation of pulmonary arterial smooth muscle cell (PASMC) phenotype is unknown. We therefore investigated in cocultured pulmonary arterial endothelial cells (PAECs) and PASMCs whether myoendothelial gap junctional signaling played a role in PAEC-dependent regulation of PASMC phenotype. Rat PAECs and PASMCs were cocultured on opposite sides of a porous Transwell membrane that permitted formation of heterotypic cell-cell contacts. Immunostaining showed expression of the gap junctional protein connexin 43 (Cx43) on projections extending into the membrane from both cell types. Dye transfer exhibited functional gap junctional communication from PAECs to PASMCs. PASMCs cocultured with PAECs had a more contractile-like phenotype (spindle shape and increased expression of the contractile proteins myosin heavy chain, H1-calponin, and α-smooth muscle cell-actin) than PASMCs cocultured with PASMCs or cocultured without direct contact with PAECs. Transforming growth factor (TGF)-ß1 signaling was activated in PASMCs cocultured with PAECs, and the PASMC differentiation was inhibited by TGF-ß type I receptor blockade. Inhibition of gap junctional communication pharmacologically or by knock down of Cx43 in PAECs blocked TGF-ß signaling and PASMC differentiation. These results implicate myoendothelial gap junctions as a gateway for PAEC-derived signals required for maintaining TGF-ß-dependent PASMC differentiation. This study identifies an alternative pathway to paracrine signaling to convey regulatory signals from PAECs to PASMCs and raises the possibility that dysregulation of this direct interaction is involved in the pathogenesis of hypertensive pulmonary vascular remodeling.


Subject(s)
Cell Communication/physiology , Connexin 43/antagonists & inhibitors , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Gap Junctions/metabolism , Lung/cytology , Lung/metabolism , Myocytes, Smooth Muscle/metabolism , Signal Transduction/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Connexin 43/metabolism , Diffusion Chambers, Culture , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Fluorescent Dyes/analysis , Gap Junctions/genetics , Gene Silencing/drug effects , Immunohistochemistry , Lung/blood supply , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
9.
J Cardiovasc Pharmacol ; 50(6): 697-702, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18091588

ABSTRACT

We have previously reported that vasoconstrictor sensitivity to KCl (a receptor-independent and voltage-gated Ca influx-mediated vasoconstrictor) is augmented in the chronically hypoxic hypertensive rat pulmonary circulation probably through increased Rho kinase-mediated Ca sensitization. However, the upstream mechanism by which the RhoA/Rho kinase signaling pathway is activated is unknown. This study examined if endogenous endothelin-1 (ET-1) and serotonin (5-HT) play roles in the Rho kinase-mediated augmented vasoconstrictor response to KCl and the activation of RhoA in chronically hypoxic hypertensive rat pulmonary arteries. The augmented KCl vasoconstriction in hypertensive lungs was reduced by the ETA receptor antagonist BQ123, while a dual ETA/B antagonist had no further effects. A combination of BQ123 and a 5-HT1B/1D receptor antagonist, GR127935, was more effective than either agent alone. The combined antagonists also reduced augmented contractile sensitivity to KCl in hypertensive intrapulmonary arteries. Membrane-to-cytosol ratio of RhoA expression in hypertensive arteries was greater than that in normotensive arteries and was reduced by BQ123 and GR127935. These results suggest that stimulation of ETA and 5-HT1B/1D receptors by endogenous ET-1 and 5-HT, respectively, is involved in RhoA/Rho kinase-mediated increased Ca sensitization in the chronically hypoxic hypertensive rat pulmonary circulation.


Subject(s)
Endothelin-1/physiology , Hypertension, Pulmonary/metabolism , Serotonin/physiology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Amides/pharmacology , Animals , Endothelin A Receptor Antagonists , Endothelin B Receptor Antagonists , Enzyme Activation , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , In Vitro Techniques , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Peptides, Cyclic/pharmacology , Perfusion/methods , Potassium Chloride/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Pulmonary Circulation/physiology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Vasoconstriction/drug effects
10.
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
11.
J Appl Physiol (1985) ; 100(3): 996-1002, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16322374

ABSTRACT

The fawn-hooded rat (FHR) develops severe pulmonary hypertension (PH) when raised for the first 3-4 wk of life in the mild hypoxia of Denver's altitude (5,280 ft.). The PH is associated with sustained pulmonary vasoconstriction and pulmonary artery remodeling. Furthermore, lung alveolarization and vascularization are reduced in the Denver FHR. We have recently shown that RhoA/Rho kinase signaling is involved in both vasoconstriction and vascular remodeling in animal models of hypoxic PH. In this study, we investigated the role of RhoA/Rho kinase signaling in the PH of Denver FHR. In alpha-toxin permeabilized pulmonary arteries from Denver FHR, the contractile sensitivity to Ca2+ was increased compared with those from sea-level FHR. RhoA activity and Rho kinase I protein expression in pulmonary arteries of Denver FHR (10-wk-old) were higher than in those of sea-level FHR. Acute inhalation of the Rho kinase inhibitor fasudil selectively reduced the elevated pulmonary arterial pressure in Denver FHR in vivo. Chronic fasudil treatment (30 mg.kg-1.day-1, from birth to 10 wk old) markedly reduced the development of PH and improved lung alveolarization and vascularization in Denver FHR. These results suggest that Rho kinase-mediated sustained vasoconstriction, through increased Ca2+ sensitivity, plays an important role in the established PH and that RhoA/Rho kinase signaling contributes significantly to the development of PH and lung dysplasia in mild hypoxia-exposed FHR.


Subject(s)
Hypertension, Pulmonary/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , rhoA GTP-Binding Protein/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Altitude , Animals , Blood Pressure/physiology , Blotting, Western , Calcium/pharmacology , Calcium/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Hemodynamics/drug effects , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Hypertrophy, Right Ventricular/metabolism , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pulmonary Alveoli/pathology , Pulmonary Artery/chemistry , Pulmonary Artery/pathology , Rats , Rats, Inbred Strains , Vasoconstriction/drug effects , rho-Associated Kinases , rhoA GTP-Binding Protein/genetics
13.
Dev Dyn ; 234(1): 1-10, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16086306

ABSTRACT

Tenascin-C (TN-C) is a mesenchyme-derived extracellular matrix (ECM) glycoprotein required for fetal lung branching morphogenesis. Given that the low oxygen (O(2)) environment of the fetus is also essential for normal lung branching morphogenesis, we determined whether fetal O(2) tension supports this process by promoting TN-C expression. Initial studies showed that 15-day fetal rat lung explants cultured for 2 days at 3% O(2) not only branched well, but they also expressed higher levels of TN-C when compared to lungs maintained at 21% O(2), which branched poorly. Antisense oligonucleotide studies demonstrated that TN-C produced in response to 3% O(2) was essential for lung branching morphogenesis. As well, exogenous TN-C protein was shown to promote branching of lung epithelial rudiments cultured at 21% O(2). Because ECM-degrading proteinases are capable of catabolizing TN-C protein, we reasoned that 3% O(2) might promote TN-C deposition by limiting the activity of these enzymes within the fetal lung. Consistent with this idea, gelatin zymography showed that the activity of a 72-kDa gelatinase, identified as matrix metalloproteinase-2 (MMP-2), was lower at 3% O(2) vs. 21% O(2). Furthermore, pharmacologic inhibition of MMP-2 activity in fetal lung explants cultured at 21% O(2) resulted in increased TN-C deposition within the mesenchyme, as well as enhanced branching morphogenesis. Collectively, these studies indicate that fetal O(2) tension promotes TN-C-dependent lung epithelial branching morphogenesis by limiting the proteolytic turnover of this ECM component within the adjacent mesenchyme.


Subject(s)
Lung/embryology , Oxygen/physiology , Tenascin/physiology , Animals , Cell Proliferation , Extracellular Matrix/physiology , Gene Expression Regulation, Developmental/physiology , Lung/physiology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mesoderm/physiology , Rats , Tissue Culture Techniques
14.
Circulation ; 111(22): 2988-96, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15927975

ABSTRACT

BACKGROUND: Human pulmonary arterial hypertension (PAH) is characterized by proliferation of vascular smooth muscle and, in its more severe form, by the development of occlusive neointimal lesions. However, few animal models of pulmonary neointimal proliferation exist, thereby limiting a complete understanding of the pathobiology of PAH. Recent studies of the endothelin (ET) system demonstrate that deficiency of the ET(B) receptor predisposes adult rats to acute and chronic hypoxic PAH, yet these animals fail to develop neointimal lesions. Herein, we determined and thereafter showed that exposure of ET(B) receptor-deficient rats to the endothelial toxin monocrotaline (MCT) leads to the development of neointimal lesions that share hallmarks of human PAH. METHODS AND RESULTS: The pulmonary hemodynamic and morphometric effects of 60 mg/kg MCT in control (MCT(+/+)) and ET(B) receptor-deficient (MCT(sl/sl)) rats at 6 weeks of age were assessed. MCT(sl/sl) rats developed more severe PAH, characterized by elevated pulmonary artery pressure, diminished cardiac output, and right ventricular hypertrophy. In MCT(sl/sl) rats, morphometric evaluation revealed the presence of neointimal lesions within small distal pulmonary arteries, increased medial wall thickness, and decreased arterial-to-alveolar ratio. In keeping with this, barium angiography revealed diminished distal pulmonary vasculature of MCT(sl/sl) rat lungs. Cells within neointimal lesions expressed smooth muscle and endothelial cell markers. Moreover, cells within neointimal lesions exhibited increased levels of proliferation and were located in a tissue microenvironment enriched with vascular endothelial growth factor, tenascin-C, and activated matrix metalloproteinase-9, factors already implicated in human PAH. Finally, assessment of steady state mRNA showed that whereas expression of ET(B) receptors was decreased in MCT(sl/sl) rat lungs, ET(A) receptor expression increased. CONCLUSIONS: Deficiency of the ET(B) receptor markedly accelerates the progression of PAH in rats treated with MCT and enhances the appearance of cellular and molecular markers associated with the pathobiology of PAH. Collectively, these results suggest an overall antiproliferative effect of the ET(B) receptor in pulmonary vascular homeostasis.


Subject(s)
Arterial Occlusive Diseases/etiology , Disease Models, Animal , Hypertension/etiology , Receptor, Endothelin B/deficiency , Tunica Intima/pathology , Animals , Animals, Genetically Modified , Cell Proliferation , Hypertension/pathology , Hypertrophy, Right Ventricular , Monocrotaline/adverse effects , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats , Receptor, Endothelin B/physiology
15.
Am J Respir Crit Care Med ; 171(5): 494-9, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15563635

ABSTRACT

We have found in chronically hypoxic rats that acute intravenous administration of the Rho kinase inhibitor Y-27632 nearly normalizes the pulmonary hypertension (PH) but has no pulmonary vascular selectivity. In this study, we tested if oral or inhaled Y-27632 would be an effective and selective pulmonary vasodilator in hypoxic PH. Although acute oral Y-27632 caused a marked and sustained decrease in mean pulmonary arterial pressure (MPAP), it also decreased mean systemic arterial pressure (MSAP). In contrast, 5 minutes of inhaled Y-27632 decreased MPAP without reducing MSAP. The hypotensive effect of inhaled Y-27632 on hypoxic PH was greater than that of inhaled nitric oxide, and the effect lasted for at least 5 hours. Inhaled fasudil, another Rho kinase inhibitor, caused selective MPAP reductions in monocrotaline-induced PH and in spontaneous PH in fawn-hooded rats, as well as in chronically hypoxic rats. These results suggested that inhaled Y-27632 was more effective than inhaled nitric oxide as a selective pulmonary vasodilator in hypoxic PH, and that Rho kinase-mediated vasoconstriction was also involved in the other models of PH. Inhaled Rho kinase inhibitors might be useful for acute vasodilator testing in patients with PH, and future work should evaluate their efficacy in the long-term treatment of PH.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Amides/administration & dosage , Enzyme Inhibitors/administration & dosage , Hypertension, Pulmonary/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/administration & dosage , Vasodilator Agents/administration & dosage , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , Administration, Inhalation , Administration, Oral , Animals , Blood Pressure/drug effects , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Hypertension, Pulmonary/etiology , Hypoxia/complications , Intracellular Signaling Peptides and Proteins , Male , Nitric Oxide/administration & dosage , Rats , Rats, Sprague-Dawley , Treatment Outcome , rho-Associated Kinases
16.
Microvasc Res ; 68(1): 1-12, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15219415

ABSTRACT

Heterogeneity in endothelial cell structure and function among vascular beds has been recognized for decades. However, recent findings have resolved that endothelial cells possess a functional memory based upon where they are in a blood vessel or based upon where they are isolated from within the blood vessel. Functional memory has been identified using integrated in vivo and in vitro bioassays and, most recently, through molecular profiling experiments. Memory is attributed to the epigenetic modification of phenotype that occurs in response to site-specific, cell-environment interaction during vascular development. In the embryo, Notch signal transduction is required for proper large blood vessel formation and function, whereas vascular endothelial cell growth factor (VEGF) is required for many of the processes of early vascular development including vasculogenesis and large vessel formation. Both Notch and VEGF are expressed in the developing lung, and their roles in pulmonary vascular development likely parallel those in the systemic circulation. Thus, integrated molecular profiling and transgenic technology provide new tools to investigate the interplay between epigenetic and environmental modulation of cell phenotype that controls endothelial cell behavior, and will aid in our understanding of the molecular signals required for normal and abnormal lung vascular development and function.


Subject(s)
Endothelial Cells/cytology , Lung/pathology , Animals , Biological Assay , Calcium/metabolism , Cell Proliferation , Cytosol/metabolism , Ethanol/pharmacology , Humans , Lung/cytology , Lung/embryology , Membrane Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Messenger/metabolism , Receptors, Notch , Signal Transduction , Thapsigargin/pharmacology , Time Factors , Transgenes , Vascular Endothelial Growth Factor A/metabolism
17.
Circ Res ; 94(11): 1507-14, 2004 Jun 11.
Article in English | MEDLINE | ID: mdl-15117820

ABSTRACT

Herein, we show that the paired-related homeobox gene, Prx1, is required for lung vascularization. Initial studies revealed that Prx1 localizes to differentiating endothelial cells (ECs) within the fetal lung mesenchyme, and later within ECs forming vascular networks. To begin to determine whether Prx1 promotes EC differentiation, fetal lung mesodermal cells were transfected with full-length Prx1 cDNA, resulting in their morphological transformation to an endothelial-like phenotype. In addition, Prx1-transformed cells acquired the ability to form vascular networks on Matrigel. Thus, Prx1 might function by promoting pulmonary EC differentiation within the fetal lung mesoderm, as well as their subsequent incorporation into vascular networks. To understand how Prx1 participates in network formation, we focused on tenascin-C (TN-C), an extracellular matrix (ECM) protein induced by Prx1. Immunocytochemistry/histochemistry showed that a TN-C-rich ECM surrounds Prx1-positive pulmonary vascular networks both in vivo and in tissue culture. Furthermore, antibody-blocking studies showed that TN-C is required for Prx1-dependent vascular network formation on Matrigel. Finally, to determine whether these results were relevant in vivo, we examined newborn Prx1-wild-type (+/+) and Prx1-null (-/-) mice and showed that Prx1 is critical for expression of TN-C and lung vascularization. These studies provide a framework to understand how Prx1 controls EC differentiation and their subsequent incorporation into functional pulmonary vascular networks.


Subject(s)
Endothelium, Vascular/cytology , Genes, Homeobox , Homeodomain Proteins/physiology , Lung/blood supply , Neovascularization, Physiologic/physiology , Tenascin/physiology , Animals , Cell Differentiation , Cell Line , Cell Movement , Endothelial Cells/cytology , Endothelium, Vascular/embryology , Extracellular Matrix/metabolism , Homeodomain Proteins/genetics , Lung/abnormalities , Lung/embryology , Mesoderm/cytology , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Tenascin/biosynthesis , Tenascin/genetics
18.
Am J Physiol Lung Cell Mol Physiol ; 287(4): L656-64, 2004 Oct.
Article in English | MEDLINE | ID: mdl-14977625

ABSTRACT

RhoA GTPase mediates a variety of cellular responses, including activation of the contractile apparatus, growth, and gene expression. Acute hypoxia activates RhoA and, in turn, its downstream effector, Rho-kinase, and previous studies in rats have suggested a role for Rho/Rho-kinase signaling in both acute and chronically hypoxic pulmonary vasoconstriction. We therefore hypothesized that activation of Rho/Rho-kinase in the pulmonary circulation of mice contributes to acute hypoxic pulmonary vasoconstriction and chronic hypoxia-induced pulmonary hypertension and vascular remodeling. In isolated, salt solution-perfused mouse lungs, acute administration of the Rho-kinase inhibitor Y-27632 (1 x 10(-5) M) attenuated hypoxic vasoconstriction as well as that due to angiotensin II and KCl. Chronic treatment with Y-27632 (30 mg x kg(-1) x day(-1)) via subcutaneous osmotic pump decreased right ventricular systolic pressure, right ventricular hypertrophy, and neomuscularization of the distal pulmonary vasculature in mice exposed to hypobaric hypoxia for 14 days. Analysis of a small number of proximal pulmonary arteries suggested that Y-27632 treatment reduced the level of phospho-CPI-17, a Rho-kinase target, in hypoxic lungs. We also found that endothelial nitric oxide synthase protein in hypoxic lungs was augmented by Y-27632, suggesting that enhanced nitric oxide production might have played a role in the Y-27632-induced attenuation of chronically hypoxic pulmonary hypertension. In conclusion, Rho/Rho-kinase activation is important in the effects of both acute and chronic hypoxia on the pulmonary circulation of mice, possibly by contributing to both vasoconstriction and vascular remodeling.


Subject(s)
Hypertension, Pulmonary/prevention & control , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pulmonary Circulation/physiology , Vasoconstriction/drug effects , Amides/therapeutic use , Animals , Cardiomegaly/physiopathology , Enzyme Inhibitors/therapeutic use , Hematocrit , Hypoxia , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Lung/drug effects , Lung/physiology , Male , Mice , Mice, Inbred C57BL , Perfusion , Pulmonary Circulation/drug effects , Pyridines/therapeutic use , Ventricular Dysfunction, Right/pathology , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right/drug effects , Ventricular Function, Right/physiology , rho-Associated Kinases
19.
Am J Physiol Lung Cell Mol Physiol ; 287(4): L665-72, 2004 Oct.
Article in English | MEDLINE | ID: mdl-12959926

ABSTRACT

Recent evidence suggests that Rho/Rho kinase signaling plays an important role in the sustained vasoconstriction induced by many agonists and is involved in the pathogenesis of systemic vascular diseases. However, little is known about its role in increased vascular tone in hypoxic pulmonary hypertension (PH). The purpose of this study was to examine whether Rho/Rho kinase-mediated Ca2+ sensitization contributed to sustained vasoconstriction and increased vasoreactivity in hypoxic PH in rats. Acute intravenous administration of Y-27632, a Rho kinase inhibitor, nearly normalized the high pulmonary arterial blood pressure and total pulmonary resistance in chronically hypoxic rats. In contrast to nifedipine, Y-27632 also markedly decreased elevated basal vascular tone in hypertensive blood-perfused lungs and isolated pulmonary arteries. Y-27632 and another Rho kinase inhibitor, HA-1077, completely reversed nitro-L-arginine-induced vasoconstriction in physiological salt solution-perfused hypertensive lungs, whereas inhibitors of myosin light chain kinase (ML-9), protein kinase C (GF-109203X), phosphatidylinositol 3-kinase (LY-294002), and tyrosine kinase (tyrphostin A23) caused only partial or no reversal of the vasoconstriction. Vasoconstrictor responses to KCl were augmented in hypertensive physiological salt solution-perfused lungs and pulmonary arteries, and the augmentation was eliminated by Y-27632. These results suggest that Rho/Rho kinase-mediated Ca2+ sensitization plays a central role in mediating sustained vasoconstriction and increased vasoreactivity in hypoxic PH.


Subject(s)
Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Protein Serine-Threonine Kinases/physiology , Pulmonary Circulation/physiology , Amides/administration & dosage , Amides/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Chromones/pharmacology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Hypertension, Pulmonary/drug therapy , Indoles/pharmacology , Injections, Intravenous , Intracellular Signaling Peptides and Proteins , Male , Maleimides/pharmacology , Morpholines/pharmacology , Nifedipine/pharmacology , Potassium Chloride/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pulmonary Circulation/drug effects , Pyridines/administration & dosage , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Vasodilator Agents/pharmacology , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL