Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 895501, 2022.
Article in English | MEDLINE | ID: mdl-35757687

ABSTRACT

Introduction: Inflammation is a major pathological feature of pulmonary arterial hypertension (PAH), particularly in the context of inflammatory conditions such as systemic sclerosis (SSc). The endothelin system and anti-endothelin A receptor (ETA) autoantibodies have been implicated in the pathogenesis of PAH, and endothelin receptor antagonists are routinely used treatments for PAH. However, immunological functions of the endothelin B receptor (ETB) remain obscure. Methods: Serum levels of anti-ETB receptor autoantibodies were quantified in healthy donors and SSc patients with or without PAH. Age-dependent effects of overexpression of prepro-endothelin-1 or ETB deficiency on pulmonary inflammation and the cardiovascular system were studied in mice. Rescued ETB-deficient mice (ETB-/-) were used to prevent congenital Hirschsprung disease. The effects of pulmonary T-helper type 2 (Th2) inflammation on PAH-associated pathologies were analyzed in ETB-/- mice. Pulmonary vascular hemodynamics were investigated in isolated perfused mouse lungs. Hearts were assessed for right ventricular hypertrophy. Pulmonary inflammation and collagen deposition were assessed via lung microscopy and bronchoalveolar lavage fluid analyses. Results: Anti-ETB autoantibody levels were elevated in patients with PAH secondary to SSc. Both overexpression of prepro-endothelin-1 and rescued ETB deficiency led to pulmonary hypertension, pulmonary vascular hyperresponsiveness, and right ventricular hypertrophy with accompanying lymphocytic alveolitis. Marked perivascular lymphocytic infiltrates were exclusively found in ETB-/- mice. Following induction of pulmonary Th2 inflammation, PAH-associated pathologies and perivascular collagen deposition were aggravated in ETB-/- mice. Conclusion: This study provides evidence for an anti-inflammatory role of ETB. ETB seems to have protective effects on Th2-evoked pathologies of the cardiovascular system. Anti-ETB autoantibodies may modulate ETB-mediated immune homeostasis.


Subject(s)
Pulmonary Arterial Hypertension , Receptor, Endothelin B , Animals , Autoantibodies/immunology , Endothelin-1/immunology , Familial Primary Pulmonary Hypertension/immunology , Humans , Hypertrophy, Right Ventricular/immunology , Inflammation/immunology , Mice , Pulmonary Arterial Hypertension/immunology , Receptor, Endothelin B/immunology , Scleroderma, Systemic/immunology
2.
Int J Mol Sci ; 21(17)2020 Sep 03.
Article in English | MEDLINE | ID: mdl-32899304

ABSTRACT

High altitude (hypobaric hypoxia) triggers several mechanisms to compensate for the decrease in oxygen bioavailability. One of them is pulmonary artery vasoconstriction and its subsequent pulmonary arterial remodeling. These changes can lead to pulmonary hypertension and the development of right ventricular hypertrophy (RVH), right heart failure (RHF) and, ultimately to death. The aim of this review is to describe the most recent molecular pathways involved in the above conditions under this type of hypobaric hypoxia, including oxidative stress, inflammation, protein kinases activation and fibrosis, and the current therapeutic approaches for these conditions. This review also includes the current knowledge of long-term chronic intermittent hypobaric hypoxia. Furthermore, this review highlights the signaling pathways related to oxidative stress (Nox-derived O2.- and H2O2), protein kinase (ERK5, p38α and PKCα) activation, inflammatory molecules (IL-1ß, IL-6, TNF-α and NF-kB) and hypoxia condition (HIF-1α). On the other hand, recent therapeutic approaches have focused on abolishing hypoxia-induced RVH and RHF via attenuation of oxidative stress and inflammatory (IL-1ß, MCP-1, SDF-1 and CXCR-4) pathways through phytotherapy and pharmacological trials. Nevertheless, further studies are necessary.


Subject(s)
Heart Failure/pathology , Hypertrophy, Right Ventricular/pathology , Hypoxia/physiopathology , Inflammation/complications , Oxidative Stress , Protein Kinases/metabolism , Animals , Heart Failure/immunology , Heart Failure/metabolism , Humans , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/metabolism , Inflammation/immunology , Inflammation/pathology
3.
J Pharmacol Exp Ther ; 349(2): 229-38, 2014 May.
Article in English | MEDLINE | ID: mdl-24563530

ABSTRACT

Airway inflammation and remodeling are major features of chronic obstructive pulmonary disease (COPD), whereas pulmonary hypertension is a common comorbidity associated with a poor disease prognosis. Recent studies in animal models have indicated that increased arginase activity contributes to features of asthma, including allergen-induced airway eosinophilia and mucus hypersecretion. Although cigarette smoke and lipopolysaccharide (LPS), major risk factors for COPD, may increase arginase expression, the role of arginase in COPD is unknown. This study aimed to investigate the role of arginase in pulmonary inflammation and remodeling using an animal model of COPD. Guinea pigs were instilled intranasally with LPS or saline twice weekly for 12 weeks and pretreated by inhalation of the arginase inhibitor 2(S)-amino-6-boronohexanoic acid (ABH) or vehicle. Repeated LPS exposure increased lung arginase activity, resulting in increased l-ornithine/l-arginine and l-ornithine/l-citrulline ratios. Both ratios were reversed by ABH. ABH inhibited the LPS-induced increases in pulmonary IL-8, neutrophils, and goblet cells as well as airway fibrosis. Remarkably, LPS-induced right ventricular hypertrophy, indicative of pulmonary hypertension, was prevented by ABH. Strong correlations were found between arginase activity and inflammation, airway remodeling, and right ventricular hypertrophy. Increased arginase activity contributes to pulmonary inflammation, airway remodeling, and right ventricular hypertrophy in a guinea pig model of COPD, indicating therapeutic potential for arginase inhibitors in this disease.


Subject(s)
Airway Remodeling , Arginase/metabolism , Pulmonary Disease, Chronic Obstructive/enzymology , Animals , Arginase/antagonists & inhibitors , Fibrosis , Guinea Pigs , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/enzymology , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/pathology , Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Lung/blood supply , Lung/enzymology , Lung/pathology , Mucin 5AC/metabolism , Neutrophils/pathology , Pneumonia/enzymology , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology
4.
Respir Res ; 14: 1, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23289668

ABSTRACT

BACKGROUND: Both chronic hypoxia and allergic inflammation induce vascular remodeling in the lung, but only chronic hypoxia appears to cause PH. We investigate the nature of the vascular remodeling and the expression and role of hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELMα) in explaining this differential response. METHODS: We induced pulmonary vascular remodeling through either chronic hypoxia or antigen sensitization and challenge. Mice were evaluated for markers of PH and pulmonary vascular remodeling throughout the lung vascular bed as well as HIMF expression and genomic analysis of whole lung. RESULTS: Chronic hypoxia increased both mean pulmonary artery pressure (mPAP) and right ventricular (RV) hypertrophy; these changes were associated with increased muscularization and thickening of small pulmonary vessels throughout the lung vascular bed. Allergic inflammation, by contrast, had minimal effect on mPAP and produced no RV hypertrophy. Only peribronchial vessels were significantly thickened, and vessels within the lung periphery did not become muscularized. Genomic analysis revealed that HIMF was the most consistently upregulated gene in the lungs following both chronic hypoxia and antigen challenge. HIMF was upregulated in the airway epithelial and inflammatory cells in both models, but only chronic hypoxia induced HIMF upregulation in vascular tissue. CONCLUSIONS: The results show that pulmonary vascular remodeling in mice induced by chronic hypoxia or antigen challenge is associated with marked increases in HIMF expression. The lack of HIMF expression in the vasculature of the lung and no vascular remodeling in the peripheral resistance vessels of the lung is likely to account for the failure to develop PH in the allergic inflammation model.


Subject(s)
Antigens , Hypertension, Pulmonary/etiology , Hypoxia/complications , Intercellular Signaling Peptides and Proteins/metabolism , Pneumonia/complications , Pulmonary Artery/metabolism , Th2 Cells/immunology , Animals , Arterial Pressure , Aspergillus/immunology , Chronic Disease , Disease Models, Animal , Familial Primary Pulmonary Hypertension , Gene Expression Profiling , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Artery/immunology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Up-Regulation
5.
Respir Res ; 12: 60, 2011 May 02.
Article in English | MEDLINE | ID: mdl-21535881

ABSTRACT

BACKGROUND: Mast cells (MCs) are implicated in inflammation and tissue remodeling. Accumulation of lung MCs is described in pulmonary hypertension (PH); however, whether MC degranulation and c-kit, a tyrosine kinase receptor critically involved in MC biology, contribute to the pathogenesis and progression of PH has not been fully explored. METHODS: Pulmonary MCs of idiopathic pulmonary arterial hypertension (IPAH) patients and monocrotaline-injected rats (MCT-rats) were examined by histochemistry and morphometry. Effects of the specific c-kit inhibitor PLX and MC stabilizer cromolyn sodium salt (CSS) were investigated in MCT-rats both by the preventive and therapeutic approaches. Hemodynamic and right ventricular hypertrophy measurements, pulmonary vascular morphometry and analysis of pulmonary MC localization/counts/activation were performed in animal model studies. RESULTS: There was a prevalence of pulmonary MCs in IPAH patients and MCT-rats as compared to the donors and healthy rats, respectively. Notably, the perivascular MCs were increased and a majority of them were degranulated in lungs of IPAH patients and MCT-rats (p < 0.05 versus donor and control, respectively). In MCT-rats, the pharmacological inhibitions of MC degranulation and c-kit with CSS and PLX, respectively by a preventive approach (treatment from day 1 to 21 of MCT-injection) significantly attenuated right ventricular systolic pressure (RVSP) and right ventricular hypertrophy (RVH). Moreover, vascular remodeling, as evident from the significantly decreased muscularization and medial wall thickness of distal pulmonary vessels, was improved. However, treatments with CSS and PLX by a therapeutic approach (from day 21 to 35 of MCT-injection) neither improved hemodynamics and RVH nor vascular remodeling. CONCLUSIONS: The accumulation and activation of perivascular MCs in the lungs are the histopathological features present in clinical (IPAH patients) and experimental (MCT-rats) PH. Moreover, the accumulation and activation of MCs in the lungs contribute to the development of PH in MCT-rats. Our findings reveal an important pathophysiological insight into the role of MCs in the pathogenesis of PH in MCT-rats.


Subject(s)
Cell Degranulation , Hypertension, Pulmonary/immunology , Lung/immunology , Mast Cells/immunology , Animals , Cell Degranulation/drug effects , Cromolyn Sodium/pharmacology , Disease Models, Animal , Familial Primary Pulmonary Hypertension , Female , Hemodynamics , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/physiopathology , Lung/blood supply , Lung/drug effects , Lung/metabolism , Male , Mast Cells/drug effects , Mast Cells/metabolism , Monocrotaline , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/metabolism , Rats , Rats, Sprague-Dawley , Ventricular Function, Right , Ventricular Remodeling
6.
Respir Res ; 12: 21, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21294880

ABSTRACT

BACKGROUND: CXCR4 is the receptor for chemokine CXCL12 and reportedly plays an important role in systemic vascular repair and remodeling, but the role of CXCR4 in development of pulmonary hypertension and vascular remodeling has not been fully understood. METHODS: In this study we investigated the role of CXCR4 in the development of pulmonary hypertension and vascular remodeling by using a CXCR4 inhibitor AMD3100 and by electroporation of CXCR4 shRNA into bone marrow cells and then transplantation of the bone marrow cells into rats. RESULTS: We found that the CXCR4 inhibitor significantly decreased chronic hypoxia-induced pulmonary hypertension and vascular remodeling in rats and, most importantly, we found that the rats that were transplanted with the bone marrow cells electroporated with CXCR4 shRNA had significantly lower mean pulmonary pressure (mPAP), ratio of right ventricular weight to left ventricular plus septal weight (RV/(LV+S)) and wall thickness of pulmonary artery induced by chronic hypoxia as compared with control rats. CONCLUSIONS: The hypothesis that CXCR4 is critical in hypoxic pulmonary hypertension in rats has been demonstrated. The present study not only has shown an inhibitory effect caused by systemic inhibition of CXCR4 activity on pulmonary hypertension, but more importantly also has revealed that specific inhibition of the CXCR4 in bone marrow cells can reduce pulmonary hypertension and vascular remodeling via decreasing bone marrow derived cell recruitment to the lung in hypoxia. This study suggests a novel therapeutic approach for pulmonary hypertension by inhibiting bone marrow derived cell recruitment.


Subject(s)
Blood Pressure , Hypertension, Pulmonary/immunology , Hypoxia/immunology , Pulmonary Artery/immunology , Receptors, CXCR4/metabolism , Animals , Benzylamines , Blood Pressure/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Transplantation , Cell Movement , Cyclams , Disease Models, Animal , Electroporation , Green Fluorescent Proteins/genetics , Heterocyclic Compounds/pharmacology , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/prevention & control , Hypoxia/drug therapy , Hypoxia/genetics , Hypoxia/pathology , Hypoxia/physiopathology , Male , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , RNA Interference , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Stem Cells/drug effects , Stem Cells/immunology , Transfection
7.
Cardiology ; 117(1): 44-53, 2010.
Article in English | MEDLINE | ID: mdl-20924177

ABSTRACT

OBJECTIVES: Thrombosis and inflammation are associated with the pathogenesis of pulmonary arterial hypertension (PAH). However, there are no solid data supporting the involvement of platelet and leukocyte activation and interaction in PAH. The present study thus investigated the activation and interaction of circulating platelets and leukocytes in a rat model of monocrotaline (MCT)-induced pulmonary hypertension. METHODS: Mean pulmonary arterial pressure (mPAP) was monitored in rats (n = 24) before and 2, 3 and 7 weeks after MCT (60 mg/kg)injection. In parallel, activation of circulating platelets and leukocytes and platelet-leukocyte aggregates were measured by whole-blood flow cytometry. RESULTS: Two weeks after MCT injection, mPAP had increased significantly, i.e. from 11.25 ± 0.92 mm Hg at baseline to 15.71 ± 1.66 mm Hg (p < 0.05), and it had increased even further at week 7 (26.83 ± 3.29 mm Hg; p < 0.01). Fibrinogen binding of circulating platelets had increased from the basal level of 1.45 ± 0.61 to 4.08 ± 1.59% 3 weeks after MCT injection (p < 0.01). Platelet responsiveness to ADP was also significantly enhanced. CD11b expression of circulating neutrophils was elevated; i.e. mean fluorescence intensity increased from 1.67 ± 0.38 before MCT injection to 2.37 ± 0.31 3 weeks after MCT injection (p < 0.01), and N-formyl-methionyl-leucyl-phenylalanine (1 × 10⁻7M) stimulation induced more marked elevation of neutrophil CD11b expression in MCT-treated animals. Circulating platelet-neutrophil aggregates were already increased 2 weeks after MCT treatment (14.93 ± 4.22%; p < 0.01) compared to baseline (6.01 ± 2.91%) and remained elevated at 3 weeks (15.19 ± 4.78%; p < 0.01). CONCLUSIONS: MCT-induced PAH in rats is associated with increased platelet and leukocyte activation and platelet-leukocyte interaction in vivo, which may play an important role in the pathogenesis of PAH.


Subject(s)
Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/physiopathology , Leukocytes/physiology , Platelet Activation/physiology , Animals , Cell Communication/immunology , Disease Models, Animal , Flow Cytometry , Hypertension, Pulmonary/chemically induced , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/physiopathology , Male , Monocrotaline , Rats , Rats, Sprague-Dawley
8.
Respir Res ; 11: 99, 2010 Jul 22.
Article in English | MEDLINE | ID: mdl-20649997

ABSTRACT

BACKGROUND: Tobacco smoking irritates and damages the respiratory tract and contributes to a higher risk of developing lung emphysema. At present, smoking cessation is the only effective treatment for reducing the progression of lung emphysema, however, there is hardly anything known about the effects of smoking cessation on cytokine and chemokine levels in the airways. To the best of our knowledge, this is the first reported in vivo study in which cytokine profiles were determined after cessation of cigarette smoke exposure. METHODS: The severity of airway remodeling and inflammation was studied by analyzing alveolar enlargement, heart hypertrophy, inflammatory cells in the bronchoalveolar lavage fluid (BALF) and lung tissue and by determining the cytokine and chemokine profiles in the BALF of A/J mice exposed to cigarette smoke for 20 weeks and 8 weeks after smoking cessation. RESULTS: The alveolar enlargement and right ventricle heart hypertrophy found in smoke-exposed mice remained unchanged after smoking cessation. Although the neutrophilic inflammation in the BALF of cigarette smoke-exposed animals was reduced after smoking cessation, a sustained inflammation in the lung tissue was observed. The elevated cytokine (IL-1 alpha and TNF-alpha) and chemokine (CCL2 and CCL3) levels in the BALF of smoke-exposed mice returned to basal levels after smoking cessation, while the increased IL-12 levels did not return to its basal level. The cigarette smoke-enhanced VEGF levels did not significantly change after smoking cessation. Moreover, IL-10 levels were reduced in the BALF of smoke-exposed mice and these levels were still significantly decreased after smoking cessation compared to the control animals. CONCLUSION: The inflammatory changes in the airways caused by cigarette smoke exposure were only partially reversed after smoking cessation. Although smoking cessation should be the first step in reducing the progression of lung emphysema, additional medication could be provided to tackle the sustained airway inflammation.


Subject(s)
Airway Remodeling , Inflammation Mediators/metabolism , Lung/immunology , Pneumonia/immunology , Smoking Cessation , Smoking/adverse effects , Animals , Bronchoalveolar Lavage Fluid/immunology , Chemokine CCL2/metabolism , Chemokine CCL3/metabolism , Disease Models, Animal , Female , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/pathology , Interleukin-1alpha/metabolism , Lung/pathology , Lung Volume Measurements , Mice , Pneumonia/pathology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology , Severity of Illness Index , Time Factors , Tumor Necrosis Factor-alpha/metabolism
9.
Respir Res ; 10: 6, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19173740

ABSTRACT

BACKGROUND: Inflammation may contribute to the pathogenesis of various forms of pulmonary hypertension (PH). Recent studies in patients with idiopathic PH or PH associated with underlying diseases suggest a role for interleukin-6 (IL-6). METHODS: To determine whether endogenous IL-6 contributes to mediate hypoxic PH and lung inflammation, we studied IL-6-deficient (IL-6-/-) and wild-type (IL-6+/+) mice exposed to hypoxia for 2 weeks. RESULTS: Right ventricular systolic pressure, right ventricle hypertrophy, and the number and media thickness of muscular pulmonary vessels were decreased in IL-6-/- mice compared to wild-type controls after 2 weeks' hypoxia, although the pressure response to acute hypoxia was similar in IL-6+/+ and IL-6-/- mice. Hypoxia exposure of IL-6+/+ mice led to marked increases in IL-6 mRNA and protein levels within the first week, with positive IL-6 immunostaining in the pulmonary vessel walls. Lung IL-6 receptor and gp 130 (the IL-6 signal transducer) mRNA levels increased after 1 and 2 weeks' hypoxia. In vitro studies of cultured human pulmonary-artery smooth-muscle-cells (PA-SMCs) and microvascular endothelial cells revealed prominent synthesis of IL-6 by PA-SMCs, with further stimulation by hypoxia. IL-6 also markedly stimulated PA-SMC migration without affecting proliferation. Hypoxic IL-6-/- mice showed less inflammatory cell recruitment in the lungs, compared to hypoxic wild-type mice, as assessed by lung protein levels and immunostaining for the specific macrophage marker F4/80, with no difference in lung expression of adhesion molecules or cytokines. CONCLUSION: These data suggest that IL-6 may be actively involved in hypoxia-induced lung inflammation and pulmonary vascular remodeling in mice.


Subject(s)
Hypertension, Pulmonary/immunology , Hypoxia/immunology , Interleukin-6/metabolism , Pneumonia/immunology , Animals , Blood Pressure , Cell Movement , Cell Proliferation , Cells, Cultured , Cytokine Receptor gp130/metabolism , Disease Models, Animal , Humans , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/immunology , Hypoxia/complications , Hypoxia/physiopathology , Interleukin-6/deficiency , Interleukin-6/genetics , Lung/immunology , Lung/pathology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , Pneumonia/physiopathology , Pulmonary Artery/immunology , Pulmonary Artery/pathology , RNA, Messenger/metabolism , Receptors, Interleukin-6/metabolism , Time Factors
10.
Circ Res ; 104(2): 236-44, 28p following 244, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19074475

ABSTRACT

Inflammatory cytokine interleukin (IL)-6 is elevated in the serum and lungs of patients with pulmonary artery hypertension (PAH). Several animal models of PAH cite the potential role of inflammatory mediators. We investigated role of IL-6 in the pathogenesis of pulmonary vascular disease. Indices of pulmonary vascular remodeling were measured in lung-specific IL-6-overexpressing transgenic mice (Tg(+)) and compared to wild-type (Tg(-)) controls in both normoxic and chronic hypoxic conditions. The Tg(+) mice exhibited elevated right ventricular systolic pressures and right ventricular hypertrophy with corresponding pulmonary vasculopathic changes, all of which were exacerbated by chronic hypoxia. IL-6 overexpression increased muscularization of the proximal arterial tree, and hypoxia enhanced this effect. It also reproduced the muscularization and proliferative arteriopathy seen in the distal arteriolar vessels of PAH patients. The latter was characterized by the formation of occlusive neointimal angioproliferative lesions that worsened with hypoxia and were composed of endothelial cells and T-lymphocytes. IL-6-induced arteriopathic changes were accompanied by activation of proangiogenic factor, vascular endothelial growth factor, the proproliferative kinase extracellular signal-regulated kinase, proproliferative transcription factors c-MYC and MAX, and the antiapoptotic proteins survivin and Bcl-2 and downregulation of the growth inhibitor transforming growth factor-beta and proapoptotic kinases JNK and p38. These findings suggest that IL-6 promotes the development and progression of pulmonary vascular remodeling and PAH through proproliferative antiapoptotic mechanisms.


Subject(s)
Blood Pressure , Hypertension, Pulmonary/immunology , Hypertrophy, Right Ventricular/immunology , Interleukin-6/metabolism , Muscle, Smooth, Vascular/immunology , Pulmonary Artery/immunology , Animals , Apoptosis , Arterioles/immunology , Arterioles/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Proliferation , Chronic Disease , Endothelial Cells/immunology , Endothelial Cells/pathology , Hyperplasia , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , Inhibitor of Apoptosis Proteins , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins c-myc/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Repressor Proteins , Survivin , Time Factors , Transforming Growth Factor beta/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Vascular Resistance , Ventricular Function, Right , Ventricular Pressure
11.
Eur Respir J ; 31(1): 135-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18166594

ABSTRACT

The pathogenesis of idiopathic pulmonary arterial hypertension (PAH) remains poorly understood. The present authors recently reported that mice with vasoactive intestinal peptide (VIP) gene disruption show a spontaneous phenotype of PAH, with pulmonary vascular remodelling and lung inflammation. To explore the underlying molecular mechanisms in this model, it was examined whether absence of the VIP gene might alter the expression of additional genes involved in the pathogenesis of PAH, as single-gene deletions, in the absence of hypoxia, rarely result in significant pulmonary vascular remodelling. Lung tissue from mice with targeted disruption of the vasoactive intestinal peptide gene (VIP(-/-) mice) and from control mice was subjected to whole-genome gene microarray analysis, and the results validated with quantitative, real-time PCR. Lungs from VIP(-/-) mice showed a wide range of significant gene expression alterations, including overexpression of genes that promote pulmonary vascular smooth muscle cell proliferation, underexpression of antiproliferative genes and upregulation of pro-inflammatory genes. In conclusion, vasoactive intestinal peptide is a pivotal modulator of genes controlling the pulmonary vasculature, its deficiency alone resulting in gene expression alterations that can readily explain both the vascular remodelling and associated inflammatory response in pulmonary arterial hypertension. The present findings shed more light on the molecular mechanisms of pulmonary arterial hypertension, and could lead to better understanding of the pathogenesis of human pulmonary arterial hypertension, and hence to improved therapy.


Subject(s)
Gene Deletion , Hypertension, Pulmonary/genetics , Hypertrophy, Right Ventricular/immunology , Vasoactive Intestinal Peptide/genetics , Animals , Humans , Hypertrophy, Right Ventricular/pathology , Hypoxia , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Vasoactive Intestinal Peptide/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 280(1): L39-49, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11133493

ABSTRACT

Tumor necrosis factor (TNF)-alpha is a key proinflammatory cytokine that is thought to be important in the development of pulmonary fibrosis, whereas its role in pulmonary emphysema has not been as thoroughly documented. In the present study, TNF-alpha was overexpressed in alveolar type II cells under the control of the human surfactant protein C promoter. In this report, we further characterized the pulmonary abnormalities and provided a physiological assessment of these mice. Histopathology of the lungs revealed chronic inflammation, severe alveolar air space enlargement and septal destruction, and bronchiolitis. However, pulmonary fibrosis was very limited and only seen in the subpleural, peribronchiolar, and perivascular regions. Physiological assessment showed an increase in lung volumes and a decrease in elastic recoil characteristic of emphysema; there was no evidence of restrictive lung disease characteristic of pulmonary fibrosis. In addition, the mice raised in ambient conditions in Denver developed pulmonary hypertension. Gelatinase activity was increased in the lavage fluid from these lungs. These results suggest that in these mice TNF-alpha contributed to the development of pulmonary emphysema through chronic lung inflammation and activation of the elastolytic enzymes but by itself was unable to produce significant pulmonary fibrosis.


Subject(s)
Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Lung Volume Measurements , Tumor Necrosis Factor-alpha/genetics , Age Factors , Altitude , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/genetics , Cytokines/immunology , Emphysema/immunology , Emphysema/pathology , Emphysema/physiopathology , Gene Expression/immunology , Hypertension, Pulmonary/immunology , Hypertrophy, Right Ventricular/immunology , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Matrix Metalloproteinase 12 , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/physiopathology , Proteolipids/genetics , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , Pulmonary Surfactants/genetics , RNA, Messenger/analysis , Respiratory Mucosa/enzymology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Th1 Cells/immunology , Transgenes/physiology , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...