Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
3.
Front Physiol ; 4: 91, 2013.
Article in English | MEDLINE | ID: mdl-23720629

ABSTRACT

Chronic Obstructive Pulmonary Disease (COPD) is one of the foremost causes of death worldwide. It is primarily caused by tobacco smoke, making it an easily preventable disease, but facilitated by genetic α-1 antitrypsin deficiency. In addition to active smokers, health problems also occur in people involuntarily exposed to second hand smoke (SHS). Currently, the relationship between SHS and COPD is not well established. Knowledge of pathogenic mechanisms is limited, thereby halting the advancement of new treatments for this socially and economically detrimental disease. Here, we attempt to summarize tobacco smoke studies undertaken in animal models, applying both mainstream (direct, nose only) and side stream (indirect, whole body) smoke exposures. This overview of 155 studies compares cellular and molecular mechanisms as well as proteolytic, inflammatory, and vasoreactive responses underlying COPD development. This is a difficult task, as listing of exposure parameters is limited for most experiments. We show that both mainstream and SHS studies largely present similar inflammatory cell populations dominated by macrophages as well as elevated chemokine/cytokine levels, such as TNF-α. Additionally, SHS, like mainstream smoke, has been shown to cause vascular remodeling and neutrophil elastase-mediated proteolytic matrix breakdown with failure to repair. Disease mechanisms and therapeutic interventions appear to coincide in both exposure scenarios. One of the more widely applied interventions, the anti-oxidant therapy, is successful for both mainstream and SHS. The comparison of direct with indirect smoke exposure studies in this review emphasizes that, even though there are many overlapping pathways, it is not conclusive that SHS is using exactly the same mechanisms as direct smoke in COPD pathogenesis, but should be considered a preventable health risk. Some characteristics and therapeutic alternatives uniquely exist in SHS-related COPD.

4.
Am J Respir Cell Mol Biol ; 48(6): 725-32, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23392573

ABSTRACT

Chronic second-hand smoke (SHS) exposure comprises the main risk factor for nonsmokers to develop chronic obstructive pulmonary disease (COPD). However, the mechanisms behind the chronic inflammation and lung destruction remain incompletely understood. In this study, we show that chronic exposure of Sprague-Dawley rats to SHS results in a significant increase of proinflammatory cytokine IL-18 and chemokine (C-C motif) ligand 5 in the bronchoalveolar lavage fluid (BALF) and a significant decrease of vascular endothelial growth factor (VEGF) in the lung tissue. SHS exposure resulted in progressive alveolar airspace enlargement, cell death, pulmonary vessel loss, vessel muscularization, collagen deposition, and right ventricular hypertrophy. Alveolar macrophages displayed a foamy phenotype and a decreased expression of the natural inhibitor of IL-18, namely, IL-18 binding protein (IL-18BP). Moreover, IL-18 down-regulated the expression of VEGF receptor-1 and VEGFR receptor-2, and induced apoptosis in pulmonary microvascular endothelial cells in vitro. We also observed a trend toward increased concentrations of IL-18 in the BALF of patients with COPD. Our findings suggest that IL-18-mediated endothelial cell death may contribute to vascular destruction and disappearance in SHS-induced COPD. Moreover, IL-18 and IL-18BP are potential new targets for therapeutics.


Subject(s)
Endothelial Cells/pathology , Interleukin-18/immunology , Pulmonary Emphysema/pathology , Tobacco Smoke Pollution/adverse effects , Animals , Bronchoalveolar Lavage Fluid/immunology , Capillary Permeability , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cell Death , Cell Line , Chemokine CCL5/immunology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Humans , Immunohistochemistry , Inhalation Exposure/adverse effects , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/immunology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Tobacco Products/adverse effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Weight Loss
5.
PLoS One ; 7(5): e38038, 2012.
Article in English | MEDLINE | ID: mdl-22675432

ABSTRACT

BACKGROUND: Given the relative abundance and toxic potential of acrolein in inhaled cigarette smoke, it is surprising how little is known about the pulmonary and systemic effects of acrolein. Here we test the hypothesis whether systemic administration of acrolein could cause endoplasmic reticulum (ER) stress, and lung cell apoptosis, leading to the enlargement of the alveolar air spaces in rats. METHODS: Acute and chronic effects of intraperitoneally administered acrolein were tested. Mean alveolar airspace area was measured by using light microscopy and imaging system software. TUNEL staining and immunohistochemistry (IHC) for active caspase 3 and Western blot analysis for active caspase 3, and caspase 12 were performed to detect apoptosis. The ER-stress related gene expression in the lungs was determined by Quantitative real-time PCR analysis. Acrolein-protein adducts in the lung tissue were detected by IHC. RESULTS: Acute administration of acrolein caused a significant elevation of activated caspase 3, upregulation of VEGF expression and induced ER stress proteins in the lung tissue. The chronic administration of acrolein in rats led to emphysematous lung tissue remodeling. TUNEL staining and IHC for cleaved caspase 3 showed a large number of apoptotic septal cells in the acrolein-treated rat lungs. Chronic acrolein administration cause the endoplasmic reticulum stress response manifested by significant upregulation of ATF4, CHOP and GADd34 expression. In smokers with COPD there was a considerable accumulation of acrolein-protein adducts in the inflammatory, airway and vascular cells. CONCLUSIONS: Systemic administration of acrolein causes endoplasmic reticulum stress response, lung cell apoptosis, and chronic administration leads to the enlargement of the alveolar air spaces and emphysema in rats. The substantial accumulation of acrolein-protein adducts in the lungs of COPD patients suggest a role of acrolein in the pathogenesis of emphysema.


Subject(s)
Acrolein/toxicity , Endoplasmic Reticulum Stress/drug effects , Pulmonary Emphysema/chemically induced , Acrolein/administration & dosage , Acrolein/metabolism , Animals , Caspase 3/metabolism , Gene Expression/drug effects , Lung/drug effects , Lung/pathology , Male , Protein Binding , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Rats , Rats, Sprague-Dawley , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , Vascular Endothelial Growth Factor A/metabolism
7.
Am J Physiol Lung Cell Mol Physiol ; 302(10): L1014-22, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22387295

ABSTRACT

In severe pulmonary arterial hypertension (PAH), vascular lesions are composed of phenotypically altered vascular and inflammatory cells that form clusters or tumorlets. Because macrophages are found in increased numbers in intravascular and perivascular space in human PAH, here we address the question whether macrophages play a role in pulmonary vascular remodeling and whether accumulation of macrophages in the lung vasculature could be compromised by the immune system. We used the mouse macrophage cell line RAW 264.7 because these cells are resistant to apoptosis, have high proliferative capacity, and resemble cells in the plexiform lesions that tend to pile up instead of maintaining a monolayer. Cells were characterized by immunocytochemistry with cell surface markers (Lycopersicon Esculentum Lectin, CD117, CD133, FVIII, CD31, VEGFR-2, and S100). Activated, but not quiescent, T cells were able to suppress RAW 264.7 cell proliferative and migration activity in vitro. The carboxyfluorescein diacetate-labeled RAW 264.7 cells were injected into the naïve Sprague Dawley (SD) rat and athymic nude rat. Twelve days later, cells were found in the lung vasculature of athymic nude rats that lack functional T cells, contributing to vascular remodeling. No labeled RAW 264.7 cells were detected in the lungs of immune-competent SD rats. Our data demonstrate that T cells can inhibit in vitro migration and in vivo accumulation of macrophage-like cells.


Subject(s)
Cell Communication/immunology , Lung/blood supply , Macrophages/cytology , Pulmonary Artery/cytology , T-Lymphocytes/cytology , Animals , Biomarkers/analysis , Cell Line , Cell Movement/immunology , Cell Proliferation , Familial Primary Pulmonary Hypertension , Fluoresceins , Fluorescent Dyes , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Immunohistochemistry , Lung/cytology , Lung/immunology , Macrophages/immunology , Macrophages/transplantation , Male , Mice , Models, Biological , Pulmonary Artery/immunology , Rats , Rats, Nude , Rats, Sprague-Dawley , T-Lymphocytes/immunology
9.
Am J Physiol Lung Cell Mol Physiol ; 302(10): L977-91, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22307907

ABSTRACT

Many chronic pulmonary diseases are associated with pulmonary hypertension (PH) and pulmonary vascular remodeling, which is a term that continues to be used to describe a wide spectrum of vascular abnormalities. Pulmonary vascular structural changes frequently increase pulmonary vascular resistance, causing PH and right heart failure. Although rat models had been standard models of PH research, in more recent years the availability of genetically engineered mice has made this species attractive for many investigators. Here we review a large amount of data derived from experimental PH reports published since 1996. These studies using wild-type and genetically designed mice illustrate the challenges and opportunities provided by these models. Hemodynamic measurements are difficult to obtain in mice, and right heart failure has not been investigated in mice. Anatomical, cellular, and genetic differences distinguish mice and rats, and pharmacogenomics may explain the degree of PH and the particular mode of pulmonary vascular adaptation and also the response of the right ventricle.


Subject(s)
Disease Models, Animal , Heart Ventricles/physiopathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/physiopathology , Pulmonary Artery/physiopathology , Animals , Blood Pressure , Familial Primary Pulmonary Hypertension , Heart Ventricles/pathology , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/pathology , Mice , Mice, Transgenic , Pulmonary Artery/pathology , Rats , Vascular Resistance , Ventricular Function, Right , Ventricular Remodeling
10.
Am J Physiol Lung Cell Mol Physiol ; 302(7): L711-8, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22307908

ABSTRACT

Efficient clearance of apoptotic cells from the lung by alveolar macrophages is important for the maintenance of tissue structure and function. Lung tissue from humans with emphysema contains increased numbers of apoptotic cells and decreased levels of vascular endothelial growth factor (VEGF). Mice treated with VEGF receptor inhibitors have increased numbers of apoptotic cells and develop emphysema. We hypothesized that VEGF regulates apoptotic cell clearance by alveolar macrophages (AM) via its interaction with VEGF receptor 1 (VEGF R1). Our data show that the uptake of apoptotic cells by murine AMs and human monocyte-derived macrophages is inhibited by depletion of VEGF and that VEGF activates Rac1. Antibody blockade or pharmacological inhibition of VEGF R1 activity also decreased apoptotic cell uptake ex vivo. Conversely, overexpression of VEGF significantly enhanced apoptotic cell uptake by AMs in vivo. These results indicate that VEGF serves a positive regulatory role via its interaction with VEGF R1 to activate Rac1 and enhance AM apoptotic cell clearance.


Subject(s)
Apoptosis , Lung/metabolism , Macrophages, Alveolar/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Antibodies, Monoclonal , Cell Line , Doxycycline/administration & dosage , Doxycycline/pharmacology , Emphysema/immunology , Emphysema/metabolism , Humans , Indoles/administration & dosage , Indoles/pharmacology , Jurkat Cells , Lung/cytology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pyrroles/administration & dosage , Pyrroles/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/immunology
11.
Respir Res ; 11: 179, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21162738

ABSTRACT

BACKGROUND: The presence of anti-endothelial cell antibodies and pathogenic T cells may reflect an autoimmune component in the pathogenesis of emphysema. Whether immune modulatory strategies can protect against the development of emphysema is not known. METHODS: Sprague Dawley rats were immunized with human umbilical vein endothelial cells (HUVEC) to induce autoimmune emphysema and treated with intrathymic HUVEC-injection and pristane. Measurements of alveolar airspace enlargement, cytokine levels, immuno histochemical, western blot analysis, and T cell repertoire of the lung tissue were performed. RESULTS: The immunomodulatory strategies protected lungs against cell death as demonstrated by reduced numbers of TUNEL and active caspase-3 positive cells and reduced levels of active caspase-3, when compared with lungs from HUVEC-immunized rats. Immunomodulatory strategies also suppressed anti-endothelial antibody production and preserved CNTF, IL-1alpha and VEGF levels. The immune deviation effects of the intrathymic HUVEC-injection were associated with an expansion of CD4+CD25+Foxp3+ regulatory T cells. Pristane treatment decreased the proportion of T cells expressing receptor beta-chain, Vß16.1 in the lung tissue. CONCLUSIONS: Our data demonstrate that interventions classically employed to induce central T cell tolerance (thymic inoculation of antigen) or to activate innate immune responses (pristane treatment) can prevent the development of autoimmune emphysema.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Autoimmunity/immunology , Cytokines/immunology , Emphysema/immunology , Emphysema/prevention & control , Immunomodulation/immunology , Animals , Male , Rats , Rats, Sprague-Dawley
12.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L954-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19684203

ABSTRACT

We recently reported that vasa vasorum expansion occurs in the pulmonary artery (PA) adventitia of chronically hypoxic animals and that extracellular ATP is a pro-angiogenic factor for isolated vasa vasorum endothelial cells (VVEC). However, the sources of extracellular ATP in the PA vascular wall, as well as the molecular mechanisms underlying its release, remain elusive. Studies were undertaken to explore whether VVEC release ATP in response to hypoxia and to determine signaling pathways involved in this process. We found that hypoxia (1-3% O2) resulted in time- and O2-dependent ATP release from VVEC. Preincubation with the inhibitors of vesicular transport (monensin, brefeldin A, and N-ethylmaleimide) significantly decreased ATP accumulation in the VVEC conditioned media, suggesting that hypoxia-induced ATP release occurs through vesicular exocytosis. Additionally, both hypoxia and exogenously added ATP resulted in the activation of PI3K and accumulation of GTP-bound RhoA in a time-dependent manner. Pharmacological inhibition of PI3K and ROCK or knockout of RhoA by small interfering RNA significantly abolished hypoxia-induced ATP release from VVEC. Moreover, RhoA and ROCK play a critical role in ATP-induced increases in VVEC DNA synthesis, migration, and tube formation, indicating a functional contribution of PI3K, Rho, and ROCK to both the autocrine mechanism of ATP release and ATP-mediated angiogenic activation of VVEC. Taken together, our findings provide novel evidence for the signaling mechanisms that link hypoxia-induced increases in extracellular ATP and vasa vasorum expansion.


Subject(s)
Adenosine Triphosphate/pharmacology , Endothelial Cells/enzymology , Neovascularization, Physiologic/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Vasa Vasorum/cytology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cattle , Cell Hypoxia/drug effects , Cell Movement/drug effects , Collagen/metabolism , DNA/biosynthesis , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Laminin/metabolism , Male , Proteoglycans/metabolism , Pulmonary Artery/cytology , Signal Transduction/drug effects , Time Factors , Transport Vesicles/drug effects , Transport Vesicles/metabolism
13.
Am J Physiol Lung Cell Mol Physiol ; 295(1): L71-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18469113

ABSTRACT

RhoA/Rho kinase (ROCK) signaling plays a key role in the pathogenesis of experimental pulmonary hypertension (PH). Dehydroepiandrosterone (DHEA), a naturally occurring steroid hormone, effectively inhibits chronic hypoxic PH, but the responsible mechanisms are unclear. This study tested whether DHEA was also effective in treating monocrotaline (MCT)-induced PH in left pneumonectomized rats and whether inhibition of RhoA/ROCK signaling was involved in the protective effect of DHEA. Three weeks after MCT injection, pneumonectomized rats developed PH with severe vascular remodeling, including occlusive neointimal lesions in pulmonary arterioles. In lungs from these animals, we detected cleaved (constitutively active) ROCK I as well as increases in activities of RhoA and ROCK and increases in ROCK II protein expression. Chronic DHEA treatment (1%, by food for 3 wk) markedly inhibited the MCT-induced PH (mean pulmonary artery pressures after treatment with 0% and 1% DHEA were 33+/-5 and 16+/-1 mmHg, respectively) and severe pulmonary vascular remodeling in pneumonectomized rats. The MCT-induced changes in RhoA/ROCK-related protein expression were nearly normalized by DHEA. A 3-wk DHEA treatment (1%) started 3 wk after MCT injection completely inhibited the progression of PH (mean pulmonary artery pressures after treatment with 0% and 1% DHEA were 47+/-3 and 30+/-3 mmHg, respectively), and this treatment also resulted in 100% survival in contrast to 30% in DHEA-untreated rats. These results suggest that inhibition of RhoA/ROCK signaling, including the cleavage and constitutive activation of ROCK I, is an important component of the impressive protection of DHEA against MCT-induced PH in pneumonectomized rats.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dehydroepiandrosterone/pharmacology , Hypertension, Pulmonary/enzymology , Monocrotaline/toxicity , rho-Associated Kinases/biosynthesis , rhoA GTP-Binding Protein/biosynthesis , Animals , Blood Pressure/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/pathology , Pneumonectomy , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Rats , Time Factors
14.
J Clin Invest ; 118(2): 394-402, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18246188

ABSTRACT

Emphysema is one manifestation of a group of chronic, obstructive, and frequently progressive destructive lung diseases. Cigarette smoking and air pollution are the main causes of emphysema in humans, and cigarette smoking causes emphysema in rodents. This review examines the concept of a homeostatically active lung structure maintenance program that, when attacked by proteases and oxidants, leads to the loss of alveolar septal cells and airspace enlargement. Inflammatory and noninflammatory mechanisms of disease pathogenesis, as well as the role of the innate and adaptive immune systems, are being explored in genetically altered animals and in exposure models of this disease. These recent scientific advances support a model whereby alveolar destruction resulting from a coalescence of mechanical forces, such as hyperinflation, and more recently recognized cellular and molecular events, including apoptosis, cellular senescence, and failed lung tissue repair, produces the clinically recognized syndrome of emphysema.


Subject(s)
Pulmonary Emphysema/etiology , Pulmonary Emphysema/metabolism , Animals , Disease Models, Animal , Guinea Pigs , Humans , Immunity, Innate , Mice , Oxidative Stress , Peptide Hydrolases/metabolism , Pulmonary Emphysema/genetics , Rats , alpha 1-Antitrypsin/metabolism
15.
J Exp Med ; 205(2): 361-72, 2008 Feb 18.
Article in English | MEDLINE | ID: mdl-18227220

ABSTRACT

Pulmonary arterial remodeling characterized by increased vascular smooth muscle density is a common lesion seen in pulmonary arterial hypertension (PAH), a deadly condition. Clinical correlation studies have suggested an immune pathogenesis of pulmonary arterial remodeling, but experimental proof has been lacking. We show that immunization and prolonged intermittent challenge via the airways with either of two different soluble antigens induced severe muscularization in small- to medium-sized pulmonary arteries. Depletion of CD4(+) T cells, antigen-specific T helper type 2 (Th2) response, or the pathogenic Th2 cytokine interleukin 13 significantly ameliorated pulmonary arterial muscularization. The severity of pulmonary arterial muscularization was associated with increased numbers of epithelial cells and macrophages that expressed a smooth muscle cell mitogen, resistin-like molecule alpha, but surprisingly, there was no correlation with pulmonary hypertension. Our data are the first to provide experimental proof that the adaptive immune response to a soluble antigen is sufficient to cause severe pulmonary arterial muscularization, and support the clinical observations in pediatric patients and in companion animals that muscularization represents one of several injurious events to the pulmonary artery that may collectively contribute to PAH.


Subject(s)
Pulmonary Artery/immunology , Pulmonary Artery/pathology , Th2 Cells/immunology , Animals , Antigens, Fungal/immunology , Aspergillus fumigatus/immunology , Blood Pressure , Heart Ventricles/physiopathology , Immunization , Interleukin-13/immunology , Interleukin-4/deficiency , Interleukin-4/genetics , Interleukin-4/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology
16.
FASEB J ; 21(13): 3640-52, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17567571

ABSTRACT

Severe pulmonary hypertension (PH) is characterized by complex precapillary arteriolar lesions, which contain phenotypically altered smooth muscle (SM) and endothelial cells (EC). We have demonstrated that VEGF receptor blockade by SU5416 {3-[(2,4-dimethylpyrrol-5-yl)methylidenyl]-indolin 2-one} in combination with chronic hypoxia causes severe angioproliferative PH associated with arterial occlusion in rats. We postulate that endothelial-mesenchymal transdifferentiation can take place in the occlusive lesions and that endothelium-derived mesenchymal cells can further differentiate toward a SM phenotype. To examine this hypothesis, we incubated human pulmonary microvascular endothelial cells (HPMVEC) with SU5416 and analyzed these cells utilizing quantitative-PCR, immunofluorescent staining and flow cytometry analysis. In vitro studies in HPMVEC demonstrated that SU5416 suppressed PGI2S gene expression while potently inducing COX-2, VEGF, and TGF-beta1 expression; and caused transdifferentiation of mature vascular endothelial cells (defined by Dil-ac-LDL, Lectin and Factor VIII) to SM-like (as defined by expression of alpha-SM actin) "transitional" cells, coexpressing both endothelial and SM markers. SU5416 expanded the number of CD34 and/or c-kit positive cells and caused transdifferentiation of CD34 positive cells but not negative cells. In conclusion, our data show that SU5416 generated a selection pressure that killed some EC and expanded progenitor-like cells to transdifferentiate to SM-like and neuronal-like cells.


Subject(s)
Antigens, CD34/immunology , Apoptosis , Cell Differentiation , Endothelium, Vascular/drug effects , Muscle, Smooth/cytology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Cell Transdifferentiation , Cells, Cultured , Fluorescent Antibody Technique , Humans
17.
Am J Respir Crit Care Med ; 175(12): 1280-9, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17413127

ABSTRACT

RATIONALE: Severe pulmonary arterial hypertension (SPH) is a frequently lethal condition characterized by pulmonary vascular remodeling and right heart strain or failure. SPH is also often associated with autoimmune and collagen vascular disorders. OBJECTIVES: To study the effects of T cells on the development of experimental SPH. METHODS: Athymic nude rats lacking T cells were treated with a single subcutaneous injection of vascular endothelial growth factor (VEGF) receptor blocker SU5416 (20 mg/kg) to induce pulmonary vascular endothelial cell apoptosis. Immunohistochemical analysis and IL-4 levels of the lung tissue were performed. Cell death and proliferation were assessed by Western blot and immunohistochemistry. MEASUREMENTS AND MAIN RESULTS: In contrast to SU5416-treated euthymic rats that develop SPH only in combination with chronic hypoxia, athymic nude rats developed SPH and vascular remodeling (similar to clinical SPH) at normoxic conditions as demonstrated by measurements of pulmonary artery pressure and right ventricle hypertrophy. Pulmonary arterioles became occluded with proliferating endothelial cells and were surrounded by mast cells, B cells, and macrophages. IL-4, proliferating cell nuclear antigen, and collagen type I levels were markedly increased in SU5416-treated athymic rat lungs. Antibody deposition was noted along the vascular endothelium in rats with SPH. Finally, protection from SPH was conferred by immune challenge with spleen cells from euthymic nude rats. CONCLUSIONS: These studies demonstrate the importance of a complete, intact immune system in protecting against pulmonary angioproliferation in this new model of SPH as well as the importance of intact VEGF receptor signaling for lung endothelial cell homeostasis.


Subject(s)
Hypertension, Pulmonary/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Angiogenesis Inhibitors , Animals , Dendritic Cells/immunology , Disease Models, Animal , Endothelial Cells , Homeostasis/immunology , Immunohistochemistry , Indoles/pharmacology , Injections, Subcutaneous , Male , Pyrroles/pharmacology , Rats , Rats, Nude , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/immunology
18.
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
20.
Am J Physiol Lung Cell Mol Physiol ; 291(4): L668-76, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16698853

ABSTRACT

Severe pulmonary hypertension (SPH) is characterized by precapillary arteriolar lumen obliteration, dramatic right ventricular hypertrophy, and pericardial effusion. Our recently published rat model of SPH recapitulates major components of the human disease. We used this model to develop new treatment strategies for SPH. SPH in rats was induced using VEGF receptor blockade in combination with chronic hypoxia. A large variety of drugs used in this study, including anticancer drugs (cyclophosphamide and paclitaxel), the angiotensin-converting enzyme inhibitor lisinopril, the antiangiogenic agent thalidomide, and the peroxisome proliferator-actived receptor-gamma agonist PGJ2, failed to decrease mean pulmonary artery pressure (PAP) or right ventricular hypertrophy. In contrast, treatment of rats with established SPH with simvastatin markedly reduced mean PAP and right ventricular hypertrophy, and this reduction was associated with caspase-3 activation and pulmonary microvascular endothelial cell apoptosis. Simvastatin partially restored caveolin-1, caveolin-2, and phospho-caveolin expression in vessel walls. In rat primary pulmonary microvascular endothelial cells, simvastatin induced caspase 3 activation and Rac 1 expression while suppressing Rho A and attenuated levels of Akt and ERK phosphorylation. We conclude that simvastatin is effective in inducing apoptosis in hyperproliferative pulmonary vascular lesions and could be considered as a potential drug for treatment of human SPH.


Subject(s)
Apoptosis/drug effects , Endothelial Cells , Endothelium, Vascular/physiopathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypertension, Pulmonary/physiopathology , Simvastatin/pharmacology , Animals , Caspase 3 , Caspases/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Enzyme Activation , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Lung/drug effects , Lung/pathology , Male , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL