Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 326(6): L786-L795, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38713613

ABSTRACT

Humans living at high-altitude (HA) have adapted to this environment by increasing pulmonary vascular and alveolar growth. RNA sequencing data from a novel murine model that mimics this phenotypical response to HA suggested estrogen signaling via estrogen receptor alpha (ERα) may be involved in this adaptation. We hypothesized ERα was a key mediator in the cardiopulmonary adaptation to chronic hypoxia and sought to delineate the mechanistic role ERα contributes to this process by exposing novel loss-of-function ERα mutant (ERαMut) rats to simulated HA. ERα mutant or wild-type (wt) rats were exposed to normoxia or hypoxia starting at conception and continued postnatally until 6 wk of age. Both wt and ERαMut animals born and raised in hypoxia exhibited lower body mass and higher hematocrits, total alveolar volumes (Va), diffusion capacities of carbon monoxide (DLCO), pulmonary arteriole (PA) wall thickness, and Fulton indices than normoxia animals. Right ventricle adaptation was maintained in the setting of hypoxia. Although no major physiologic differences were seen between wt and ERαMut animals at either exposure, ERαMut animals exhibited smaller mean linear intercepts (MLI) and increased PA total and lumen areas. Hypoxia exposure or ERα loss-of-function did not affect lung mRNA abundance of vascular endothelial growth factor, angiopoietin 2, or apelin. Sexual dimorphisms were noted in PA wall thickness and PA lumen area in ERαMut rats. In summary, in room air-exposed rats and rats with peri- and postnatal hypoxia exposure, ERα loss-of-function was associated with decreased alveolar size (primarily driven by hypoxic animals) and increased PA remodeling.NEW & NOTEWORTHY By exposing novel loss-of-function estrogen receptor alpha (Erα) mutant rats to a novel model of human high-altitude exposure, we demonstrate that ERα has subtle but inconsistent effects on endpoints relevant to cardiopulmonary adaptation to chronic hypoxia. Given that we observed some histologic, sex, and genotype differences, further research into cell-specific effects of ERα during hypoxia-induced cardiopulmonary adaptation is warranted.


Subject(s)
Adaptation, Physiological , Estrogen Receptor alpha , Hypoxia , Animals , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Female , Hypoxia/metabolism , Hypoxia/physiopathology , Rats , Male , Lung/metabolism , Lung/pathology , Altitude , Disease Models, Animal , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics
2.
J Clin Invest ; 131(6)2021 03 15.
Article in English | MEDLINE | ID: mdl-33497359

ABSTRACT

Women with pulmonary arterial hypertension (PAH) exhibit better right ventricular (RV) function and survival than men; however, the underlying mechanisms are unknown. We hypothesized that 17ß-estradiol (E2), through estrogen receptor α (ER-α), attenuates PAH-induced RV failure (RVF) by upregulating the procontractile and prosurvival peptide apelin via a BMPR2-dependent mechanism. We found that ER-α and apelin expression were decreased in RV homogenates from patients with RVF and from rats with maladaptive (but not adaptive) RV remodeling. RV cardiomyocyte apelin abundance increased in vivo or in vitro after treatment with E2 or ER-α agonist. Studies employing ER-α-null or ER-ß-null mice, ER-α loss-of-function mutant rats, or siRNA demonstrated that ER-α is necessary for E2 to upregulate RV apelin. E2 and ER-α increased BMPR2 in pulmonary hypertension RVs and in isolated RV cardiomyocytes, associated with ER-α binding to the Bmpr2 promoter. BMPR2 is required for E2-mediated increases in apelin abundance, and both BMPR2 and apelin are necessary for E2 to exert RV-protective effects. E2 or ER-α agonist rescued monocrotaline pulmonary hypertension and restored RV apelin and BMPR2. We identified what we believe to be a novel cardioprotective E2/ER-α/BMPR2/apelin axis in the RV. Harnessing this axis may lead to novel RV-targeted therapies for PAH patients of either sex.


Subject(s)
Apelin/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Hypertension, Pulmonary/physiopathology , Ventricular Function, Right/physiology , Animals , Cardiotonic Agents/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Female , Humans , Male , Mice , Mice, Knockout , Models, Cardiovascular , Myocytes, Cardiac/metabolism , Rats , Rats, Mutant Strains
3.
Am J Physiol Lung Cell Mol Physiol ; 315(3): L382-L386, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29745251

ABSTRACT

We have shown that cigarette smoke (CS)-induced pulmonary emphysema-like manifestations are preceded by marked suppression of the number and function of bone marrow hematopoietic progenitor cells (HPCs). To investigate whether a limited availability of HPCs may contribute to CS-induced lung injury, we used a Food and Drug Administration-approved antagonist of the interactions of stromal cell-derived factor 1 (SDF-1) with its chemokine receptor CXCR4 to promote intermittent HPC mobilization and tested its ability to limit emphysema-like injury following chronic CS. We administered AMD3100 (5mg/kg) to mice during a chronic CS exposure protocol of up to 24 wk. AMD3100 treatment did not affect either lung SDF-1 levels, which were reduced by CS, or lung inflammatory cell counts. However, AMD3100 markedly improved CS-induced bone marrow HPC suppression and significantly ameliorated emphysema-like end points, such as alveolar airspace size, lung volumes, and lung static compliance. These results suggest that antagonism of SDF-1 binding to CXCR4 is associated with protection of both bone marrow and lungs during chronic CS exposure, thus encouraging future studies of potential therapeutic benefit of AMD3100 in emphysema.


Subject(s)
Heterocyclic Compounds/pharmacology , Lung Injury , Pulmonary Alveoli/metabolism , Pulmonary Emphysema , Smoking , Animals , Benzylamines , Bone Marrow/metabolism , Bone Marrow/pathology , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/metabolism , Cyclams , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Lung Injury/etiology , Lung Injury/metabolism , Lung Injury/pathology , Lung Injury/prevention & control , Mice , Pulmonary Alveoli/pathology , Pulmonary Emphysema/etiology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Pulmonary Emphysema/prevention & control , Receptors, CXCR4/metabolism , Smoking/adverse effects , Smoking/metabolism , Smoking/pathology
4.
Am J Respir Cell Mol Biol ; 59(1): 114-126, 2018 07.
Article in English | MEDLINE | ID: mdl-29394091

ABSTRACT

17ß-Estradiol (E2) attenuates hypoxia-induced pulmonary hypertension (HPH) through estrogen receptor (ER)-dependent effects, including inhibition of hypoxia-induced endothelial cell proliferation; however, the mechanisms responsible for this remain unknown. We hypothesized that the protective effects of E2 in HPH are mediated through hypoxia-inducible factor 1α (HIF-1α)-dependent increases in ERß expression. Sprague-Dawley rats and ERα or ERß knockout mice were exposed to hypobaric hypoxia for 2-3 weeks. The effects of hypoxia were also studied in primary rat or human pulmonary artery endothelial cells (PAECs). Hypoxia increased expression of ERß, but not ERα, in lungs from HPH rats as well as in rat and human PAECs. ERß mRNA time dependently increased in PAECs exposed to hypoxia. Normoxic HIF-1α/HIF-2α stabilization increased PAEC ERß, whereas HIF-1α knockdown decreased ERß abundance in hypoxic PAECs. In turn, ERß knockdown in hypoxic PAECs increased HIF-2α expression, suggesting a hypoxia-sensitive feedback mechanism. ERß knockdown in hypoxic PAECs also decreased expression of the HIF inhibitor prolyl hydroxylase 2 (PHD2), whereas ERß activation increased PHD2 and decreased both HIF-1α and HIF-2α, suggesting that ERß regulates the PHD2/HIF-1α/HIF-2α axis during hypoxia. Whereas hypoxic wild-type or ERα knockout mice treated with E2 demonstrated less pulmonary vascular remodeling and decreased HIF-1α after hypoxia compared with untreated hypoxic mice, ERß knockout mice exhibited increased HIF-2α and an attenuated response to E2 during hypoxia. Taken together, our results demonstrate a novel and potentially therapeutically targetable mechanism whereby hypoxia, via HIF-1α, increases ERß expression and the E2-ERß axis targets PHD2, HIF-1α, and HIF-2α to attenuate HPH development.


Subject(s)
Endothelial Cells/metabolism , Estrogen Receptor beta/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/pathology , Pulmonary Artery/pathology , Up-Regulation , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/drug effects , Cells, Cultured , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/agonists , Estrogen Receptor beta/metabolism , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Lung/pathology , Male , Nitriles/pharmacology , Procollagen-Proline Dioxygenase/metabolism , Propionates/pharmacology , Protein Stability/drug effects , Rats, Sprague-Dawley , Up-Regulation/drug effects , Vascular Remodeling/drug effects
5.
Pulm Circ ; 7(1): 232-243, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28680582

ABSTRACT

17ß-estradiol (E2) exerts complex and context-dependent effects in pulmonary hypertension. In hypoxia-induced pulmonary hypertension (HPH), E2 attenuates lung vascular remodeling through estrogen receptor (ER)-dependent effects; however, ER target genes in the hypoxic lung remain unknown. In order to identify the genome regulated by the E2-ER axis in the hypoxic lung, we performed a microarray analysis in lungs from HPH rats treated with E2 (75 mcg/kg/day) ± ER-antagonist ICI182,780 (3 mg/kg/day). Untreated HPH rats and normoxic rats served as controls. Using a false discovery rate of 10%, we identified a significantly differentially regulated genome in E2-treated versus untreated hypoxia rats. Genes most upregulated by E2 encoded matrix metalloproteinase 8, S100 calcium binding protein A8, and IgA Fc receptor; genes most downregulated by E2 encoded olfactory receptor 63, secreted frizzled-related protein 2, and thrombospondin 2. Several genes affected by E2 changed in the opposite direction after ICI182,780 co-treatment, indicating an ER-regulated genome in HPH lungs. The bone morphogenetic protein antagonist Grem1 (gremlin 1) was upregulated by hypoxia, but found to be among the most downregulated genes after E2 treatment. Gremlin 1 protein was reduced in E2-treated versus untreated hypoxic animals, and ER-blockade abolished the inhibitory effect of E2 on Grem1 mRNA and protein. In conclusion, E2 ER-dependently regulates several genes involved in proliferative and inflammatory processes during hypoxia. Gremlin 1 is a novel target of the E2-ER axis in HPH. Understanding the mechanisms of E2 gene regulation in HPH may allow for selectively harnessing beneficial transcriptional activities of E2 for therapeutic purposes.

6.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L375-88, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27288487

ABSTRACT

17ß-Estradiol (E2) exerts protective effects on right ventricular (RV) function in pulmonary arterial hypertension (PAH). Since acute exercise-induced increases in afterload may lead to RV dysfunction in PAH, we sought to determine whether E2 allows for superior RV adaptation after an acute exercise challenge. We studied echocardiographic, hemodynamic, structural, and biochemical markers of RV function in male and female rats with sugen/hypoxia (SuHx)-induced pulmonary hypertension, as well as in ovariectomized (OVX) SuHx females, with or without concomitant E2 repletion (75 µg·kg(-1)·day(-1)) immediately after 45 min of treadmill running at 75% of individually determined maximal aerobic capacity (75% aerobic capacity reserve). Compared with males, intact female rats exhibited higher stroke volume and cardiac indexes, a strong trend for better RV compliance, and less pronounced increases in indexed total pulmonary resistance. OVX abrogated favorable RV adaptations, whereas E2 repletion after OVX markedly improved RV function. E2's effects on pulmonary vascular remodeling were complex and less robust than its RV effects. Postexercise hemodynamics in females with endogenous or exogenous E2 were similar to hemodynamics in nonexercised controls, whereas OVX rats exhibited more severely altered postexercise hemodynamics. E2 mediated inhibitory effects on RV fibrosis and attenuated increases in RV collagen I/III ratio. Proapoptotic signaling, endothelial nitric oxide synthase phosphorylation, and autophagic flux markers were affected by E2 depletion and/or repletion. Markers of impaired autophagic flux correlated with endpoints of RV structure and function. Endogenous and exogenous E2 exerts protective effects on RV function measured immediately after an acute exercise challenge. Harnessing E2's mechanisms may lead to novel RV-directed therapies.


Subject(s)
Estradiol/physiology , Hypertension, Pulmonary/physiopathology , Adaptation, Physiological , Animals , Arterial Pressure , Autophagy , Estradiol/pharmacology , Female , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/physiopathology , Male , Nitric Oxide Synthase Type III/metabolism , Oxygen Consumption , Physical Exertion , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , Sex Characteristics , Stroke Volume , Vascular Remodeling , Ventricular Dysfunction, Right , Ventricular Function, Right , Ventricular Pressure
7.
Am J Physiol Lung Cell Mol Physiol ; 308(8): L797-806, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25659904

ABSTRACT

The development of pulmonary hypertension (PH) requires multiple pulmonary vascular insults, yet the role of early oxygen therapy as an initial pulmonary vascular insult remains poorly defined. Here, we employ a two-hit model of PH, utilizing postnatal hyperoxia followed by adult hypoxia exposure, to evaluate the role of early hyperoxic lung injury in the development of later PH. Sprague-Dawley pups were exposed to 90% oxygen during postnatal days 0-4 or 0-10 or to room air. All pups were then allowed to mature in room air. At 10 wk of age, a subset of rats from each group was exposed to 2 wk of hypoxia (Patm = 362 mmHg). Physiological, structural, and biochemical endpoints were assessed at 12 wk. Prolonged (10 days) postnatal hyperoxia was independently associated with elevated right ventricular (RV) systolic pressure, which worsened after hypoxia exposure later in life. These findings were only partially explained by decreases in lung microvascular density. Surprisingly, postnatal hyperoxia resulted in robust RV hypertrophy and more preserved RV function and exercise capacity following adult hypoxia compared with nonhyperoxic rats. Biochemically, RVs from animals exposed to postnatal hyperoxia and adult hypoxia demonstrated increased capillarization and a switch to a fetal gene pattern, suggesting an RV more adept to handle adult hypoxia following postnatal hyperoxia exposure. We concluded that, despite negative impacts on pulmonary artery pressures, postnatal hyperoxia exposure may render a more adaptive RV phenotype to tolerate late pulmonary vascular insults.


Subject(s)
Hyperoxia/physiopathology , Lung Injury/physiopathology , Animals , Animals, Newborn , Cell Hypoxia , Female , Hyperoxia/complications , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/metabolism , Lung Injury/etiology , Male , Rats, Sprague-Dawley , Ventricular Remodeling
8.
Am J Physiol Lung Cell Mol Physiol ; 308(9): L873-90, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25713318

ABSTRACT

Estrogens are disease modifiers in PAH. Even though female patients exhibit better right ventricular (RV) function than men, estrogen effects on RV function (a major determinant of survival in PAH) are incompletely characterized. We sought to determine whether sex differences exist in RV function in the SuHx model of PAH, whether hormone depletion in females worsens RV function, and whether E2 repletion improves RV adaptation. Furthermore, we studied the contribution of ERs in mediating E2's RV effects. SuHx-induced pulmonary hypertension (SuHx-PH) was induced in male and female Sprague-Dawley rats as well as OVX females with or without concomitant E2 repletion (75 µg·kg(-1)·day(-1)). Female SuHx rats exhibited superior CI than SuHx males. OVX worsened SuHx-induced decreases in CI and SuHx-induced increases in RVH and inflammation (MCP-1 and IL-6). E2 repletion in OVX rats attenuated SuHx-induced increases in RV systolic pressure (RVSP), RVH, and pulmonary artery remodeling and improved CI and exercise capacity (V̇o2max). Furthermore, E2 repletion ameliorated SuHx-induced alterations in RV glutathione activation, proapoptotic signaling, cytoplasmic glycolysis, and proinflammatory cytokine expression. Expression of ERα in RV was decreased in SuHx-OVX but was restored upon E2 repletion. RV ERα expression was inversely correlated with RVSP and RVH and positively correlated with CO and apelin RNA levels. RV-protective E2 effects observed in females were recapitulated in male SuHx rats treated with E2 or with pharmacological ERα or ERß agonists. Our data suggest significant RV-protective ER-mediated effects of E2 in a model of severe PH.


Subject(s)
Estradiol/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/physiopathology , Animals , Apelin , Apoptosis , Blood Pressure/drug effects , Body Weight/drug effects , Cytokines/biosynthesis , Estrogen Receptor alpha/biosynthesis , Female , Inflammation , Intercellular Signaling Peptides and Proteins/genetics , Male , Ovariectomy , Ovary/surgery , Physical Conditioning, Animal , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Sex Factors , Vascular Remodeling , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects
9.
Pulm Circ ; 4(2): 300-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25006449

ABSTRACT

Endothelin-1 is a potent mediator of sepsis-induced pulmonary hypertension (PH). The pulmonary vascular effects of selective blockade of endothelin receptor subtype A (ETAR) during endotoxemia remain unknown. We hypothesized that selective ETAR antagonism attenuates endotoxin-induced PH and improves pulmonary artery (PA) vasoreactivity. Adult male Sprague-Dawley rats (250-450 g) received lipopolysaccharide (LPS; Salmonella typhimurium; 20 mg/kg intraperitoneally) or vehicle 6 hours before hemodynamic assessment and tissue harvest. The selective ETAR antagonist sitaxsentan (10 or 20 mg/kg) or vehicle was injected intravenously 3 hours after receipt of LPS. Right ventricular systolic pressure, mean arterial pressure (MAP), cardiac output (CO), oxygenation (P/F ratio), and serum bicarbonate were measured. Bronchoalveolar lavage (BAL) cell differential and lung wet-to-dry ratios were obtained. Endothelium-dependent and endothelium-independent vasorelaxations were determined in isolated PA rings. PA interleukin (IL)-1ß, IL-6, tumor necrosis factor α (TNF-α), and inducible nitric oxide synthase (iNOS) messenger RNA (mRNA) were measured. LPS caused PH, decreased MAP, CO, and serum bicarbonate, and increased PA IL-1ß, IL-6, TNF-α, and iNOS mRNA. Sitaxsentan attenuated sepsis-induced PH and increased MAP. The P/F ratio, CO, serum bicarbonate, and BAL neutrophilia were not affected by sitaxsentan. In isolated PA rings, while not affecting phenylephrine-induced vasocontraction or endothelium-dependent relaxation, sitaxsentan dose-dependently attenuated LPS-induced alterations in endothelium-independent relaxation. PA cytokine mRNA levels were not significantly attenuated by ETAR blockade. We conclude that ETAR blockade attenuates endotoxin-induced alterations in systemic and PA pressures without negatively affecting oxygenation. This protective effect appears to be mediated not by attenuation of sepsis-induced cardiac dysfunction, acidosis, or alveolar inflammation but rather by improved endothelium-independent vasorelaxation.

10.
J Bone Miner Res ; 29(2): 361-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23873717

ABSTRACT

Fibroblast growth factor 23 (FGF23) gain of function mutations can lead to autosomal dominant hypophosphatemic rickets (ADHR) disease onset at birth, or delayed onset following puberty or pregnancy. We previously demonstrated that the combination of iron deficiency and a knock-in R176Q FGF23 mutation in mature mice induced FGF23 expression and hypophosphatemia that paralleled the late-onset ADHR phenotype. Because anemia in pregnancy and in premature infants is common, the goal of this study was to test whether iron deficiency alters phosphate handling in neonatal life. Wild-type (WT) and ADHR female breeder mice were provided control or iron-deficient diets during pregnancy and nursing. Iron-deficient breeders were also made iron replete. Iron-deficient WT and ADHR pups were hypophosphatemic, with ADHR pups having significantly lower serum phosphate (p < 0.01) and widened growth plates. Both genotypes increased bone FGF23 mRNA (>50 fold; p < 0.01). WT and ADHR pups receiving low iron had elevated intact serum FGF23; ADHR mice were affected to a greater degree (p < 0.01). Iron-deficient mice also showed increased Cyp24a1 and reduced Cyp27b1, and low serum 1,25-dihydroxyvitamin D (1,25D). Iron repletion normalized most abnormalities. Because iron deficiency can induce tissue hypoxia, oxygen deprivation was tested as a regulator of FGF23, and was shown to stimulate FGF23 mRNA in vitro and serum C-terminal FGF23 in normal rats in vivo. These studies demonstrate that FGF23 is modulated by iron status in young WT and ADHR mice and that hypoxia independently controls FGF23 expression in situations of normal iron. Therefore, disturbed iron and oxygen metabolism in neonatal life may have important effects on skeletal function and structure through FGF23 activity on phosphate regulation.


Subject(s)
Familial Hypophosphatemic Rickets/metabolism , Fibroblast Growth Factors/metabolism , Iron Deficiencies , Mutation, Missense , Phosphates/metabolism , Amino Acid Substitution , Animals , Animals, Newborn , Calcitriol/genetics , Calcitriol/metabolism , Diet/adverse effects , Familial Hypophosphatemic Rickets/genetics , Familial Hypophosphatemic Rickets/pathology , Female , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Gene Knock-In Techniques , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/pathology , Iron/pharmacology , Male , Mice, Mutant Strains , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Vitamin D3 24-Hydroxylase
11.
Am J Respir Crit Care Med ; 185(9): 965-80, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22383500

ABSTRACT

RATIONALE: 17ß-Estradiol (E2) attenuates hypoxic pulmonary vasoconstriction and hypoxic pulmonary hypertension (HPH) through an unknown mechanism that may involve estrogen receptors (ER) or E2 conversion to catecholestradiols and methoxyestradiols with previously unrecognized effects on cardiopulmonary vascular remodeling. OBJECTIVES: To determine the mechanism by which E2 exerts protective effects in HPH. METHODS: Male rats were exposed to hypobaric hypoxia while treated with E2 (75 µg/kg/d) or vehicle. Subgroups were cotreated with pharmacologic ER-antagonist or with inhibitors of E2-metabolite conversion. Complementary studies were performed in rats cotreated with selective ERα- or ERß-antagonist. Hemodynamic and pulmonary artery (PA) and right ventricular (RV) remodeling parameters, including cell proliferation, cell cycle, and autophagy, were measured in vivo and in cultured primary rat PA endothelial cells. MEASUREMENTS AND MAIN RESULTS: E2 significantly attenuated HPH endpoints. Hypoxia increased ERß but not ERα lung vascular expression. Co-treatment with nonselective ER inhibitor or ERα-specific antagonist rendered hypoxic animals resistant to the beneficial effects of E2 on cardiopulmonary hemodynamics, whereas ERα- and ERß-specific antagonists opposed the remodeling effects of E2. In contrast, inhibition of E2-metabolite conversion did not abolish E2 protection. E2-treated hypoxic animals exhibited reduced ERK1/2 activation and increased expression of cell-cycle inhibitor p27(Kip1) in lungs and RV, with up-regulation of lung autophagy. E2-induced signaling was recapitulated in hypoxic but not normoxic endothelial cells, and was associated with decreased vascular endothelial growth factor secretion and cell proliferation. CONCLUSIONS: E2 attenuates hemodynamic and remodeling parameters in HPH in an ER-dependent manner, through direct antiproliferative mechanisms on vascular cells, which may provide novel nonhormonal therapeutic targets for HPH.


Subject(s)
Estradiol/pharmacology , Hypertension, Pulmonary/drug therapy , Hypoxia/complications , Receptors, Estrogen/drug effects , Airway Remodeling/drug effects , Airway Remodeling/physiology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cardiac Output/drug effects , Cardiac Output/physiology , Cyclin-Dependent Kinase Inhibitor p27/drug effects , Cyclin-Dependent Kinase Inhibitor p27/physiology , Estradiol/analogs & derivatives , Estradiol/therapeutic use , Estrogen Antagonists/pharmacology , Fulvestrant , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypoxia/drug therapy , Hypoxia/physiopathology , Lung/blood supply , Lung/physiopathology , Male , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/physiology , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
12.
Lung Cancer ; 63(3): 341-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18614255

ABSTRACT

Smoking causes lung cancer and chronic obstructive pulmonary disease (COPD) that impose severe health problem to humans. Both diseases are related to each other and can be induced by chronic inflammation in the lung. To identify the molecular mechanism for lung cancer formation, a CCSP-rtTA/(teto)(7)Stat3C bitransgenic model was generated recently. In this model, persistent activation of the Stat3 signaling pathway induced pulmonary inflammation and adenocarcinoma formation in the lung. A group of Stat3 downstream genes were identified by Affymetrix GeneChip microarray analysis that can be used as biomarkers for lung cancer diagnosis and prognosis. To determine which human lung cancers are related to the Stat3 pathway, multiple Stat3 downstream genes were screened in human lung cancers (adenocarcinomas and squamous cell carcinomas) and lung tissue with COPD. In both cancer and COPD, the Stat3 gene was up-regulated. A panel of Stat3-up-regulated downstream genes in mice was up-regulated in human adenocarcinomas, but not in human squamous cell carcinomas. This panel of genes was also modestly up-regulated in lung tissue with COPD from patients with a history of smoking and not up-regulated in those without histories of smoking. Several Stat3-down-regulated downstream genes also showed differential expression patterns in carcinoma and COPD. These studies support a concept that Stat3 is a potent oncogenic molecule that plays a role in formation of lung adenocarcinomas in both mice and humans. The carcinogenesis of adenocarcinoma and squamous cell carcinoma is mediated by different molecular mechanisms and pathways in vivo. Stat3 and its downstream genes can serve as biomarkers for lung adenocarcinoma and COPD diagnosis and prognosis in mice and humans.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Pulmonary Disease, Chronic Obstructive/genetics , RNA, Neoplasm/genetics , STAT3 Transcription Factor/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Biomarkers/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Polymerase Chain Reaction , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , STAT3 Transcription Factor/biosynthesis
13.
J Rheumatol ; 32(2): 325-34, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15693095

ABSTRACT

OBJECTIVE: To determine if intraarticular (IA) injection of hyaluronan (HA) into the canine knee after anterior cruciate ligament transection (ACLT) alters the progression of osteoarthritis (OA) and the perception of pain in this model. METHODS: OA was induced in 30 adult dogs of mixed breed by ACLT. The dogs were divided into 3 groups and given 5 weekly IA injections into the unstable knee during Weeks 1-5 and 13-17. The prophylactic treatment group received HA in the first series and saline during the second series. The therapeutic group received saline in the first series and HA in the second series. The control group received saline during both injection series. The progression of joint damage of OA was evaluated by arthroscopy 12 weeks after ACLT and by gross examination 32 weeks after ACLT. Histologic and biochemical changes of OA were evaluated. Loading of the unstable limb during gait was determined by force-plate analysis before surgery, after each series of injections, and the week before euthanasia. RESULTS: Arthroscopic examination 12 weeks after ACLT revealed OA changes in the cruciate-deficient knees. Chondropathy scores ranged from 0 to 8 (possible range 0-65). The mean chondropathy score was 2.5 +/- 1.3 (mean +/- SD) for the controls, 2.5 +/- 2.5 for the therapeutic group, and 2.1 +/- 1.3 for the prophylactic group. At the termination of the experiment 32 weeks after ACLT, the gross chondropathy scores were 14.0 +/- 5.2 for controls, 17.6 +/- 6.8 for the therapeutic group, and 13.3 +/- 5.0 for the prophylactic group. There were no significant differences among the means of the gross scores, the histologic scores, or biochemical composition of articular cartilage. The peak vertical ground reaction force (VGRF) generated by the unstable limb was reduced by ~60% after ACLT, and slowly increased to ~75% of the baseline value over the 32 weeks after ACLT. HA injection had no effect on the VGRF or on the pathologic changes of OA. CONCLUSION: Intraarticular HA injection did not alter the progression of OA in the cruciate-deficient canine knee or alter the loading of the unstable limb.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Anterior Cruciate Ligament Injuries , Hyaluronic Acid/therapeutic use , Joint Instability/drug therapy , Osteoarthritis, Knee/drug therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Anterior Cruciate Ligament/surgery , Arthroscopy , Biomechanical Phenomena , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Disease Models, Animal , Disease Progression , Dogs , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/pharmacology , Injections, Intra-Articular , Joint Instability/pathology , Joint Instability/physiopathology , Osteoarthritis, Knee/physiopathology , Stifle/drug effects , Stifle/pathology
14.
Am J Ther ; 3(1): 17-20, 1996 Jan.
Article in English | MEDLINE | ID: mdl-11856993

ABSTRACT

The ability of misoprostol to reverse the deleterious changes induced in cartilage by sodium salicylate was tested using osteoarthritic canine and chondrocytes. Adult mongrel dogs were subjected to anterior cruciate ligament transection and dosed with either misoprostol, salicylate, or misoprostol plus salicylate. No significant differences were noted among the three groups in either gross and histological changes or general biochemical changes. This approach was abandoned since the levels of misoprostol attained by oral dosing were much lower than those required for domonstration of misoprostol effects in vitro. Next, chondrocytes were isolated from the osteoarthritic and contralateral knee joint cartilage of dogs 12 weeks after anterior cruciate ligament transection and cultured in alginate beads. The cultures were incubated with (3)H-proline and both the genetic types of collagen synthesized and the net synthesis of (3)H-hydroxyproline were determined. No drug or combination of drugs affected the genetic types of collagen synthesized. Total protein and collagen synthesis by chondrocytes was reduced in the presence of salicylate and increased in the presence of misoprostol. When osteoarthritic chondrocytes were incubated with both agents, the salicylate effect was reversed by misoprostol. Collagen synthesis by the chondrocytes from the contralateral knee was also suppressed by salicylate, but addition of misoprostol failed to restore synthesis. In summary, misoprostol, even at very high doses, has limited chondroprotective activity in canine cartilage, as judged from collagen synthetic activity.

SELECTION OF CITATIONS
SEARCH DETAIL
...