Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Dtsch Med Wochenschr ; 141(S 01): S4-S9, 2016 Oct.
Article in German | MEDLINE | ID: mdl-27760444

ABSTRACT

The 2015 European Guidelines on Diagnosis and Treatment of Pulmonary Hypertension (PH) are also valid for Germany. While the guidelines contain detailed recommendations regarding clinical aspects of pulmonary arterial hypertension (PAH) and other forms of PH, they contain only a relatively short paragraph on novel findings on the pathobiology, pathology, and genetics. However, these are of great importance for our understanding of this complex disease both from a clinical and scientific point of view, and they are essential for the development of novel treatment strategies. To this end, a number of current data are relevant, prompting a detailed commentary to the guidelines, and the consideration of new scientific data. In June 2016, a Consensus Conference organized by the PH working groups of the German Society of Cardiology (DGK), the German Society of Respiratory Medicine (DGP) and the German Society of Pediatric Cardiology (DGPK) was held in Cologne, Germany. This conference aimed to solve practical and controversial issues surrounding the implementation of the European Guidelines in Germany. To this end, a number of working groups was initiated, one of which was specifically dedicated to the pathobiology, pathology and genetics of PH. This article summarizes the results and recommendations of this working group.


Subject(s)
Cardiology/standards , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/therapy , Practice Guidelines as Topic , Pulmonary Medicine/standards , Antihypertensive Agents/therapeutic use , Combined Modality Therapy/standards , Endarterectomy/standards , Germany , Humans , Hypertension, Pulmonary/genetics
2.
Pneumologie ; 69(3): 135-43, 2015 Mar.
Article in German | MEDLINE | ID: mdl-25750094

ABSTRACT

Riociguat is the first clinically available soluble Guanylate-cyclase stimulator (sGC) and representative of a completely new class of drugs. Riociguat is approved for pulmonary arterial hypertension (PAH) and non-operable or recurrent/persistent chronic thromboembolic pulmonary hypertension (CTEPH). Moreover, Riociguat is currently under investigation for a wider spectrum of diseases. This article focusses on its mode of action and clinical trial data. Finally, based on these data, the status of approval, as well as the costs a proposal is given how Riociguat can be integrated in the current treatment of PAH and CTEPH.


Subject(s)
Guanylate Cyclase/metabolism , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Pulmonary Embolism/drug therapy , Pulmonary Embolism/metabolism , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Receptors, Cytoplasmic and Nuclear/metabolism , Antihypertensive Agents/administration & dosage , Chronic Disease , Fibrinolytic Agents/administration & dosage , Humans , Hypertension, Pulmonary/complications , Pulmonary Embolism/complications , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Receptors, Cytoplasmic and Nuclear/agonists , Soluble Guanylyl Cyclase , Treatment Outcome
3.
Thromb Haemost ; 112(6): 1288-303, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25231004

ABSTRACT

Chronic exposure to hypoxia induces a pronounced remodelling of the pulmonary vasculature leading to pulmonary hypertension (PH). The remodelling process also entails increased proliferation and decreased apoptosis of pulmonary arterial smooth muscle cells (PASMC), processes regulated by the cytoskeletal protein paxillin. In this study, we aimed to examine the molecular mechanisms leading to deregulation of paxillin in PH. We detected a time-dependent increase in paxillin tyrosine 31 (Y31) and 118 (Y118) phosphorylation following hypoxic exposure (1 % O2) or platelet-derived growth factor (PDGF)-BB stimulation of primary human PASMC. In addition, both, hypoxia- and PDGF-BB increased the nuclear localisation of phospho-paxillin Y31 as indicated by immunofluorescence staining in human PASMC. Elevated paxillin tyrosine phosphorylation in human PASMC was attenuated by hypoxia-inducible factor (HIF)-1α depletion or by treatment with the PDGF-BB receptor antagonist, imatinib. Moreover, we observed elevated paxillin Y31 and Y118 phosphorylation in the pulmonary vasculature of chronic hypoxic mice (21 days, 10 % O2) which was reversible by imatinib-treatment. PDGF-BB-dependent PASMC proliferation was regulated via the paxillin-Erk1/2-cyclin D1 pathway. In conclusion, we suggest paxillin up-regulation and phosphorylation as an important mechanism of vascular remodelling underlying pulmonary hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Benzamides/pharmacology , Hypertension, Pulmonary/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Paxillin/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-sis/pharmacology , Pyrimidines/pharmacology , Vascular Remodeling/drug effects , Active Transport, Cell Nucleus , Animals , Apoptosis/drug effects , Becaplermin , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Imatinib Mesylate , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Paxillin/genetics , Phosphorylation , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , RNA Interference , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Tyrosine
4.
Br J Pharmacol ; 171(16): 3895-907, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24779394

ABSTRACT

BACKGROUND AND PURPOSE: Pulmonary arteries (PAs) are innervated, but little is known about the role of neuronal axis in pulmonary hypertension (PH). Here, we have examined the role of the neuropeptide Y (NPY) and its Y1 receptor in PH pathogenesis. EXPERIMENTAL APPROACH: NPY was localized by immunofluorescence. Expression of NPY and Y1 receptor were determined by quantitative PCR. Cellular response to NPY stimulation was assessed by Western blotting, thymidine incorporation and calcium imaging. Wire myography and isolated perfused mouse lung were applied to study pulmonary vasoactive effects of NPY. Selective receptor antagonists were used to assess the contribution of receptor subtypes in mediating NPY effects. KEY RESULTS: Samples from PH patients showed increased NPYergic innervation within the PA wall and higher Y1 receptor expression, compared with donors. However, NPY levels were unchanged in both PA and serum. In the chronic hypoxic mouse model, Y1 receptor were up-regulated, while expression of both NPY and Y1 receptor was increased in the lungs of monocrotaline and SU5416-hypoxia rats. On a functional level, NPY acutely increased intracellular calcium levels and enhanced vasoconstriction of lung vessels preconstricted with adrenaline. Furthermore, NPY stimulated proliferation of human pulmonary arterial smooth muscle cells and activated p38 and PKD pathways. Correspondingly, higher phosphorylation of PKD was observed in remodelled vessels from PH patients. The selective Y1 receptor antagonist, BIBO 3304, concentration-dependently inhibited vasoconstrictive and proliferative effects of NPY. CONCLUSIONS AND IMPLICATIONS: NPY and Y1 receptor are possible mediators of both vasoconstriction and pulmonary vascular remodelling in PH.


Subject(s)
Hypertension, Pulmonary/physiopathology , Neuropeptide Y/physiology , Receptors, Neuropeptide Y/physiology , Adult , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cell Proliferation/drug effects , Epinephrine/pharmacology , Female , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Hypoxia/physiopathology , In Vitro Techniques , Indoles , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Male , Mice, Inbred C57BL , Monocrotaline , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Pyrroles , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Young Adult
5.
Oncogene ; 32(9): 1121-34, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-22525277

ABSTRACT

Lung cancer is the leading cause of cancer death worldwide. Recent data suggest that cyclic nucleotide phosphodiesterases (PDEs) are relevant in various cancer pathologies. Pathophysiological role of phosphodiesterase 4 (PDE4) with possible therapeutic prospects in lung cancer was investigated. We exposed 10 different lung cancer cell lines (adenocarcinoma, squamous and large cell carcinoma) to hypoxia and assessed expression and activity of PDE4 by real-time PCR, immunocytochemistry, western blotting and PDE activity assays. Expression and activity of distinct PDE4 isoforms (PDE4A and PDE4D) increased in response to hypoxia in eight of the studied cell lines. Furthermore, we analyzed various in silico predicted hypoxia-responsive elements (p-HREs) found in in PDE4A and PDE4D genes. Performing mutation analysis of the p-HRE in luciferase reporter constructs, we identified four functional HRE sites in the PDE4A gene and two functional HRE sites in the PDE4D gene that mediated hypoxic induction of the reporter. Silencing of hypoxia-inducible factor subunits (HIF1α and HIF2α) by small interfering RNA reduced hypoxic induction of PDE4A and PDE4D. Vice versa, using a PDE4 inhibitor (PDE4i) as a cyclic adenosine monophosphate (cAMP) -elevating agent, cAMP analogs or protein kinase A (PKA)-modulating drugs and an exchange protein directly activated by cAMP (EPAC) activator, we demonstrated that PDE4-cAMP-PKA/EPAC axis enhanced HIF signaling as measured by HRE reporter gene assay, HIF and HIF target genes expression ((lactate dehydrogenase A), LDHA, (pyruvate dehydrogenase kinase 1) PDK1 and (vascular endothelial growth factor A) VEGFA). Notably, inhibition of PDE4 by PDE4i or silencing of PDE4A and PDE4D reduced human lung tumor cell proliferation and colony formation. On the other hand, overexpression of PDE4A or PDE4D increased human lung cancer proliferation. Moreover, PDE4i treatment reduced hypoxia-induced VEGF secretion in human cells. In vivo, PDE4i inhibited tumor xenograft growth in nude mice by attenuating proliferation and angiogenesis. Our findings suggest that PDE4 is expressed in lung cancer, crosstalks with HIF signaling and promotes lung cancer progression. Thus, PDE4 may represent a therapeutic target for lung cancer therapy.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Humans , Mice , Mice, Nude , Phosphodiesterase 4 Inhibitors/pharmacology , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Transplantation, Heterologous
6.
Intensive Care Med ; 37(8): 1368-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21626431

ABSTRACT

PURPOSE: The calcium-sensitizing drug levosimendan increases myocardial contractility and, by activating K(+)-channels, dilates pulmonary vessels. In the acute setting, levosimendan is clinically used to treat right heart failure in pulmonary hypertension. As K(+)-channel activation elicits several beneficial effects in the vascular system, we hypothesized that levosimendan also attenuates the remodeling process in the monocrotaline model of rat pulmonary hypertension. METHODS AND RESULTS: Animal subgroups received levosimendan, the K(+)-channel opener nicorandil, or levosimendan together with the K(+)-adenosine triphosphate (ATP)-sensitive potassium channel (K(ATP)) blocker glibenclamide. Morphometric analyses revealed that levosimendan and nicorandil attenuated the increased pulmonary vascular medial wall thickness after monocrotaline challenge. Accordingly, in vivo BrdU assays revealed that levosimendan significantly diminished proliferation of pulmonary arterial smooth muscle cells (PASMCs), and this effect was attenuated by glibenclamide. Levosimendan also reduced right ventricular hypertrophy, but this effect was not glibenclamide sensitive and not recapitulated by nicorandil. In cell culture, levosimendan had a direct inhibitory effect on the platelet-derived growth factor (PDGF)-induced proliferation of PASMCs, which however required high concentrations of the compound, pointing towards an endothelial effect. Indeed, levosimendan increased cyclic guanosine monophosphate (cGMP) in human umbilical vein endothelial cells (HUVECs) and impaired the tumor necrosis factor-α (TNF-α)-induced inflammatory expression of E-selectin, intercellular adhesion molecule-1 (ICAM-1), cyclooxygenase-2 (COX-2), and monocyte chemotactic protein-1 (MCP-1). In luciferase reporter gene assays in HUVECs, levosimendan dose-dependently attenuated the TNF-α-stimulated increase of proinflammatory transcription factors activator protein 1 (AP1), hypoxia-inducible factor-1α (HIF-1α), and nuclear factor-κB (NF-κB). CONCLUSIONS: Levosimendan attenuates pulmonary vascular remodeling, presumably by an antiproliferative and anti-inflammatory effect which is mediated by cellular hyperpolarization. The compound also has a direct inhibitory effect on cardiac hypertrophy, which is however K(+)-channel independent.


Subject(s)
Airway Remodeling/drug effects , Hydrazones/pharmacology , Hypertension, Pulmonary/drug therapy , Pulmonary Circulation/drug effects , Pyridazines/pharmacology , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Disease Models, Animal , Germany , Glyburide/administration & dosage , Glyburide/pharmacology , Hydrazones/administration & dosage , Hypertension, Pulmonary/chemically induced , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Myocardial Contraction/drug effects , Nicorandil/administration & dosage , Nicorandil/pharmacology , Potassium Channels/drug effects , Pyridazines/administration & dosage , Rats , Rats, Sprague-Dawley , Simendan , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacology
7.
Br J Pharmacol ; 163(8): 1721-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21410457

ABSTRACT

BACKGROUND AND PURPOSE Arachidonic acid derivatives play a central role in inflammation processes. Arachidonic acid is metabolized by several enzymes, particularly cyclooxygenases (COX), 5-lipoxygenase (5-LOX) and microsomal prostaglandin E-synthase-1 (mPGES-1) to pro-inflammatory mediators. EXPERIMENTAL APPROACH We determined the effect of LP105, a pirinixic acid derivative which acts as inhibitor of 5-LOX, COX and mPGES-1, on aortic aneurysm development in mice and on 5-LOX activity in murine monocytes. KEY RESULTS In a monocyte cell line (RAW264.7), LP105 inhibited 5-LOX in whole cells (IC(50) : 1-3 µM) and in supernatants (IC(50) : ∼10 µM). Oral administration of LP105 to mice resulted in therapeutic tissue and plasma levels. Aortic aneurysms were induced in ApoE(-/-) mice by angiotensin II (AngII) and LP105 (5 mg·day(-1) per animal) was co-administered to a subgroup. Compared with animals receiving AngII alone, the LP105+AngII group showed a lower heart rate, a trend towards reduced heart to body weight ratio but similar hypertensive responses. AngII alone significantly increased aortic weight and diameter but co-treatment with LP105+AngII prevented these changes. LC/MS-MS studies revealed increased 15-hydroxytetraenoic acid (15-HETE) and 14,15-epoxyeicosatrienoic acid (14,15-EET) plasma levels in LP105-treated animals. In the murine kidney, mRNAs of EET-generating or metabolizing enzymes and of 5-LOX and 15-LOX were unaffected by LP105. LP105 also did not inhibit the EET-metabolizing soluble epoxide hydrolase. CONCLUSIONS AND IMPLICATIONS LP105 was a potent inhibitor of monocyte 5-LOX and reduced AngII-induced vascular remodelling in mice. A shift of arachidonic acid metabolism to the protective EET pathway may contribute to the beneficial effects of LP105.


Subject(s)
Aortic Aneurysm/pathology , Arachidonate 5-Lipoxygenase/metabolism , Cardiotonic Agents/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Lipoxygenase Inhibitors/pharmacology , Pyrimidines/pharmacology , Administration, Oral , Angiotensin II/administration & dosage , Angiotensin II/toxicity , Animals , Aorta/pathology , Aortic Aneurysm/metabolism , Arachidonate 5-Lipoxygenase/blood , Cardiotonic Agents/pharmacokinetics , Cardiotonic Agents/therapeutic use , Cardiovascular System/drug effects , Cell Line , Cyclooxygenase Inhibitors/therapeutic use , Disease Models, Animal , Epoxide Hydrolases/blood , Epoxide Hydrolases/metabolism , Injections, Subcutaneous , Lipoxygenase Inhibitors/therapeutic use , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prostaglandin-E Synthases , Pyrimidines/metabolism , Pyrimidines/therapeutic use
8.
Eur Respir J ; 37(5): 1104-18, 2011 May.
Article in English | MEDLINE | ID: mdl-20947677

ABSTRACT

Pulmonary arterial hypertension (PAH) is a life-threatening disease characterised by vasoconstriction and remodelling of the pulmonary vasculature. The serotonin (5-hydroxytryptamine (5-HT)) pathway has been shown to play a major role in the pathogenesis of PAH, but pharmacological modulation of this pathway for treatment of PAH is, to date, at a pre-clinical level. Terguride is a 5-HT receptor (5-HTR) antagonist that is well tolerated and clinically approved for ovulation disorders. Immunohistochemistry against 5-HTR(2A/B) on human lungs revealed their localisation to the vascular smooth muscle layer and quantitative RT-PCR showed 5-HTR(2B) upregulation in pulmonary artery smooth muscle cells (PASMC) isolated from PAH patients. Proliferation and migration of cultured primary human PASMC were dose-dependently blocked by terguride. Therapeutic 5-HT signalling inhibition was 1) demonstrated in isolated, ventilated and perfused rat lungs and 2) by chronic terguride treatment of rats with monocrotaline (MCT)-induced pulmonary hypertension in a preventive or curative approach. Terguride inhibited proliferation of PASMCs and abolished 5-HT-induced pulmonary vasoconstriction. Chronic terguride treatment prevented dose-dependently the development and progression of MCT-induced PAH in rats. Thus, terguride represents a valuable novel therapeutic approach in PAH.


Subject(s)
Dopamine Agonists/therapeutic use , Hypertension, Pulmonary/drug therapy , Lisuride/analogs & derivatives , Lung/drug effects , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Adult , Animals , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/pathology , Lisuride/therapeutic use , Lung/pathology , Lung/physiopathology , Lung Transplantation , Male , Monocrotaline/pharmacology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats
9.
Clin Microbiol Infect ; 17(1): 7-14, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20545963

ABSTRACT

Inflammation underlies a wide variety of physiological and pathological processes. Acute inflammation is the initial response of the body to harmful stimuli. Chronic inflammation, by contrast, is a prolonged, dysregulated and maladaptive response that involves active inflammation, tissue destruction and attempts at tissue repair. Over the past few years, such persistent inflammation has been shown to be associated with pulmonary hypertension (PH). Substantial advances in basic and experimental science have illuminated the role of inflammation and the underlying cellular and molecular mechanisms that contribute to PH. This review summarizes the experimental and clinical evidence for inflammation in various types of PH. In addition, it assesses the current state of knowledge regarding the inducers/triggers of chronic inflammation and infection, as well as the inflammatory mediators and cells that are involved in PH. Infiltration of inflammatory cells, such as dendritic cells, macrophages, mast cells, T-lymphocytes and B-lymphocytes, in the vascular lesions and an elevation of serum/tissue concentrations of proinflammatory cytokines and chemokines and their contribution to pulmonary vascular remodelling are reported in detail. We review the data supporting the use of inflammatory markers as prognostic and predictive factors in PH. Finally, we consider how new insights into inflammation in PH may identify innovative therapeutic strategies.


Subject(s)
Communicable Diseases/complications , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/immunology , Pneumonia/immunology , Adaptive Immunity , Animals , B-Lymphocytes/immunology , Humans , Hypertension, Pulmonary/physiopathology , Immunity, Innate , Inflammation Mediators/immunology , Pneumonia/complications , T-Lymphocytes/immunology
10.
Eur Respir J ; 37(6): 1400-10, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21148228

ABSTRACT

Left heart disease (LHD) frequently causes lung vascular remodelling and pulmonary hypertension (PH). Yet pharmacological treatment for PH in LHD is lacking and its pathophysiological basis remains obscure. We aimed to identify candidate mechanisms of PH in LHD and to test their relevance and therapeutic potential. In rats, LHD was induced by supracoronary aortic banding. Whole genome microarray analyses were performed, candidate genes were confirmed by RT-PCR and Western blots and functional relevance was tested in vivo by genetic and pharmacological strategies. In lungs of LHD rats, mast cell activation was the most prominently upregulated gene ontology cluster. Mast cell gene upregulation was confirmed at RNA and protein levels and remodelled vessels showed perivascular mast cell accumulations. In LHD rats treated with the mast cell stabiliser ketotifen, or in mast cell deficient Ws/Ws rats, PH and vascular remodelling were largely attenuated. Both strategies also reduced PH and vascular remodelling in monocrotaline-induced pulmonary arterial hypertension, suggesting that the role of mast cells extends to non-cardiogenic PH. In PH of different aetiologies, mast cells accumulate around pulmonary blood vessels and contribute to vascular remodelling and PH. Mast cells and mast cell-derived mediators may present promising targets for the treatment of PH.


Subject(s)
Hypertension, Pulmonary/etiology , Lung/blood supply , Mast Cells/physiology , Ventricular Dysfunction, Left/complications , Animals , Gene Expression Profiling , Histamine H1 Antagonists/pharmacology , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Ketotifen/pharmacology , Lung/metabolism , Male , Monocrotaline/toxicity , Rats , Rats, Sprague-Dawley , Up-Regulation , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism
11.
Eur Respir J ; 36(6): 1302-14, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20525716

ABSTRACT

A unique subpopulation of peripheral blood mononuclear cells that exhibit a parallel expression of haematopoietic and mesenchymal markers has been described as "circulating fibrocytes". These cells were demonstrated to obtain a fibroblastic phenotype in tissues or cell culture and contribute to pulmonary fibrotic disorders and tissue remodelling processes. The aim of our study was to characterise the recruitment of circulating fibrocytes in vivo in the model of chronic hypoxic pulmonary hypertension in mice and to analyse the therapeutic effect of the stable prostacyclin analogue trepostinil with respect to this cell population. To track circulating fibrocytes in vivo, we transplanted wild-type mice with bone marrow from ubiquitously eGFP expressing mice and subjected them to chronic hypoxia. We observed significantly increased recruitment of circulating fibrocytes to the remodelled pulmonary resistance arteries in response to hypoxia. Treatment with treprostinil significantly reduced the recruitment of these cells compared to normoxic mice. Treprostinil also reduced right ventricular systolic pressure and slightly reduced the vascular remodelling but failed to reverse the right ventricular hypertrophy. In summary, we show that circulating fibrocytes contribute to hypoxic pulmonary vascular remodelling and may be specifically targeted by a prostacyclin analogue. Further investigations of cellular and paracrine mechanisms are warranted to decipher their role in pulmonary hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Antihypertensive Agents/pharmacokinetics , Antihypertensive Agents/therapeutic use , Epoprostenol/analogs & derivatives , Fibroblasts/drug effects , Hypertension, Pulmonary/drug therapy , Leukocytes, Mononuclear/drug effects , Animals , Blood Circulation , Blood Vessels/drug effects , Blood Vessels/physiopathology , Bone Marrow Cells/drug effects , Chimerism , Chronic Disease , Epoprostenol/pharmacology , Epoprostenol/therapeutic use , Female , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/physiopathology , Lung/blood supply , Lung/drug effects , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology
12.
Eur Respir J ; 36(4): 808-18, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20530035

ABSTRACT

An accumulating body of evidence incriminates Rho kinase (ROCK) in the pathogenesis of pulmonary hypertension (PH). The therapeutic efficacy of azaindole-1, a novel highly selective and orally active ROCK inhibitor, has not yet been investigated in PH. This study aimed to investigate the effects of azaindole-1 on 1) acute hypoxic pulmonary vasoconstriction (HPV), 2) proliferation of pulmonary arterial smooth muscle cells (PASMCs) and 3) animal models of PH. Azaindole-1 significantly inhibited HPV in isolated, ventilated and buffer-perfused murine lungs and proliferation of primary rat PASMCs in vitro. Azaindole-1 was administered orally from 21 to 35 days after monocrotaline (MCT) injection in rats and hypoxic exposure in mice. Azaindole-1 (10 and 30 mg per kg body weight per day in rats and mice, respectively) significantly improved haemodynamics and right ventricular hypertrophy. Moreover, the medial wall thickness and muscularisation of peripheral pulmonary arteries were ameliorated. Azaindole-1 treatment resulted in a decreased immunoreactivity for phospho-myosin phosphatase target subunit 1 and proliferating cell nuclear antigen in pulmonary vessels of MCT-injected rats, suggesting an impaired ROCK activity and reduced proliferating cells. Azaindole-1 provided therapeutic benefit in experimental PH, and this may be attributable to its potent vasorelaxant and antiproliferative effects. Azaindole-1 may offer a useful approach for treatment of PH.


Subject(s)
Azabicyclo Compounds/therapeutic use , Hypertension, Pulmonary/drug therapy , Indoles/therapeutic use , Animals , Cell Proliferation , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hemodynamics , Lung/pathology , Male , Mice , Rats , Rats, Sprague-Dawley , Telemetry/methods , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Thymidine/chemistry , Treatment Outcome , rho-Associated Kinases/antagonists & inhibitors
13.
Eur Respir J ; 36(5): 1056-66, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20516051

ABSTRACT

Hypoxic pulmonary vasoconstriction (HPV) is an essential mechanism to optimise lung gas exchange. We aimed to decipher the proposed oxygen sensing mechanism of mitochondria in HPV. Cytochrome redox state was assessed by remission spectrophotometry in intact lungs and isolated pulmonary artery smooth muscle cells (PASMC). Mitochondrial respiration was quantified by high-resolution respirometry. Alterations were compared with HPV and hypoxia-induced functional and molecular readouts on the cellular level. Aortic and renal arterial smooth muscle cells (ASMC and RASMC, respectively) served as controls. The hypoxia-induced decrease of mitochondrial respiration paralleled HPV in isolated lungs. In PASMC, reduction of respiration and mitochondrial cytochrome c and aa3 (complex IV), but not of cytochrome b (complex III) matched an increase in matrix superoxide levels as well as mitochondrial membrane hyperpolarisation with subsequent cytosolic calcium increase. In contrast to PASMC, RASMC displayed a lower decrease in respiration and no rise in superoxide, membrane potential or intracellular calcium. Pharmacological inhibition of mitochondria revealed analogous kinetics of cytochrome redox state and strength of HPV. Our data suggest inhibition of complex IV as an essential step in mitochondrial oxygen sensing of HPV. Concomitantly, increased superoxide release from complex III and mitochondrial membrane hyperpolarisation may initiate the cytosolic calcium increase underlying HPV.


Subject(s)
Cytochromes/metabolism , Hypoxia/metabolism , Lung/metabolism , Mitochondria/metabolism , Muscle, Smooth, Vascular/metabolism , Oxygen Consumption/physiology , Animals , Aorta/cytology , Cell Respiration/physiology , Cells, Cultured , Cytochromes b/metabolism , Cytochromes c/metabolism , Electron Transport Complex IV/metabolism , Female , Lung/blood supply , Male , Membrane Potential, Mitochondrial/physiology , Muscle, Smooth, Vascular/cytology , Oxidation-Reduction , Pulmonary Circulation/physiology , Rabbits , Renal Artery/cytology , Spectrophotometry , Superoxides/metabolism , Vasoconstriction/physiology
14.
Eur Respir J ; 36(1): 187-95, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20032014

ABSTRACT

Anti-neutrophil cytoplasmic antibodies (c-ANCA) targeting proteinase 3 (PR3) are implicated in the pathogenesis of Wegener's granulomatosis (WG). Fulminant disease can present as acute lung injury (ALI). In this study, a model of ALI in WG was developed using isolated rat lungs. Isolated human polymorphonuclear leukocytes (PMNs) were primed with tumour necrosis factor (TNF) to induce surface expression of PR3. Co-perfusion of TNF-primed neutrophils and monoclonal anti-PR3 antibodies induced a massive weight gain in isolated lungs. This effect was not observed when control immunoglobulin G was co-perfused with TNF-primed PMNs. The c-ANCA-induced oedema formation was paralleled by an increase in the capillary filtration coefficient as a marker of increased pulmonary endothelial permeability. In contrast, pulmonary artery pressure was not affected. In the presence of the oxygen radical scavenger superoxide dismutase and a NADPH oxidase inhibitor, c-ANCA-induced lung oedema could be prevented. Inhibition of neutrophil elastase was equally effective in preventing c-ANCA-induced lung injury. In conclusion, anti-PR3 antibodies induced neutrophil mediated, elastase- and oxygen radical-dependent ALI in the isolated lung. This experimental model supports the hypothesis of a pathogenic role for c-ANCA in WG and offers the possibility of the development of therapeutic strategies for the treatment of lung injury in fulminant WG.


Subject(s)
Acute Lung Injury/immunology , Antibodies, Antineutrophil Cytoplasmic/immunology , Granulomatosis with Polyangiitis/immunology , Neutrophils/immunology , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Antibodies, Antineutrophil Cytoplasmic/pharmacology , Antibodies, Monoclonal/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Granulomatosis with Polyangiitis/drug therapy , Granulomatosis with Polyangiitis/prevention & control , Humans , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Leukocyte Elastase/antagonists & inhibitors , Myeloblastin/immunology , NADPH Oxidases/antagonists & inhibitors , Neutrophil Activation/immunology , Pulmonary Edema/immunology , Pulmonary Edema/prevention & control , Rats , Superoxide Dismutase/analysis , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
15.
Dtsch Med Wochenschr ; 134 Suppl 5: S157-9, 2009 Aug.
Article in German | MEDLINE | ID: mdl-19718604

ABSTRACT

Pulmonary arterial hypertension is a life-threatening, vasculoproliferative disease of the lung, which is characterized by vasoconstriction and remodeling of small pulmonary arteries. Drugs for the treatment of PAH mainly address the increased vascular tone. Substances like prostacyclin, endothelin-receptor-antagonists and phosphodiesterase-5-inhibitors have been approved for the treatment of PAH and represent the current therapeutic options. The development of a causal treatment aiming a normalization of the vessel wall structure is the current focus of research. The key events in disease progression are represented by increased proliferation, migration and a resistance to apoptosis of pulmonary vascular cells. Therefore, new non-vasoactive drugs are investigated in relevant preclinical animal models of pulmonary arterial hypertension. Some of these substances, like tyrosine kinase inhibitors, elastase inhibitors and phosphodiesterase-1-inhibitors, could not only attenuate (anti-remodeling) but reverse (reverse-remodeling) the disease. Additionally, new vasodilators, like soluble guanylate cyclase stimulators and activators, addressing well-known and new signaling pathways are currently under investigation. Taken together, with increasing insight into the pathology of PAH, several novel drug targets and treatments have emerged which may improve the management of patients and which efficacy is currently addressed in preclinical studies and clinical trials.


Subject(s)
Hypertension, Pulmonary/drug therapy , Pulmonary Artery/physiopathology , Animals , Disease Progression , Endothelin Receptor Antagonists , Epoprostenol/pharmacology , Epoprostenol/therapeutic use , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypertension, Pulmonary/pathology , NADPH Oxidases/metabolism , Pancreatic Elastase/antagonists & inhibitors , Phosphodiesterase 5 Inhibitors , Phosphodiesterase I/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Reactive Oxygen Species/metabolism , Serotonin Antagonists/therapeutic use , Signal Transduction/drug effects , Vasoconstriction/drug effects , Vasodilator Agents/therapeutic use , rho-Associated Kinases/antagonists & inhibitors
16.
Eur J Clin Invest ; 39 Suppl 2: 3-13, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19335741

ABSTRACT

Pulmonary hypertension (PH), a chronic disorder of the pulmonary vasculature, is characterized by progressive elevation in pulmonary artery pressure and the ultimate development of right-sided heart failure and death. Being a rapidly progressive disease with limited therapeutic options, the pathogenesis of PH is complex and multifactorial. The pathogenesis may result from a combination of vasoconstriction, inward vascular wall remodelling and in situ thrombosis that involves dysfunction of underlying cellular pathways and mediators. Among these, the activation of endothelin (ET) system has been shown to be important in the development and perpetuation of PH. Endothelin-1 (ET-1), a potent vasoconstrictor and mitogen, exerts its biological effects by binding to two G-protein-coupled receptor isoforms, endothelin A (ETA) receptor and endothelin B (ETB) receptor. These two receptors are nonredundant and unique because of distinct localization, unique binding locations and affinities for the endothelin peptide and activation of distinct signalling pathways. Importantly, there is now substantial evidence that direct antagonism of ET receptors that can block either ETA- or ETA- and ETB receptors can be beneficial for the treatment of PH in both preclinical and clinical setting. This review provides an overview of endothelin biology, various preclinical models that have been widely used to investigate the pathophysiology of PH as well as the individual roles of the ET receptors (ETA and ETB) and their regulation in disease pathogenesis. We also review current data on the use of selective and nonselective ET receptor antagonism in the preclinical PH models.


Subject(s)
Antihypertensive Agents/therapeutic use , Disease Models, Animal , Endothelin Receptor Antagonists , Hypertension, Pulmonary/drug therapy , Animals , Drug Evaluation, Preclinical/methods , Endothelins/physiology , Humans , Hypertension, Pulmonary/physiopathology
17.
Eur J Clin Invest ; 39 Suppl 2: 38-49, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19335746

ABSTRACT

Pathological vascular remodelling is a key contributor to the symptomatology of pulmonary arterial hypertension (PAH), and reversing this process may offer the best hope for improving this debilitating condition. The vascular remodelling process is believed to be due to endothelial cell dysfunction and to involve altered production of endothelial cell-derived vasoactive mediators. The observation that circulating plasma levels of the vasoactive peptide endothelin (ET)-1 are raised in patients with PAH, and that ET-1 production is increased in the pulmonary tissue of affected individuals, makes it a particularly interesting target for a therapeutic intervention in PAH. Clinical trials with ET receptor antagonists (ETRAs) show that they provide symptomatic benefit in patients with PAH, thereby proving the clinical relevance of the ET system as a therapeutic target. In this paper, we review the role of ET-1 together with the available data on the roles of the specific ET receptors and ETRAs in PAH. In particular, we discuss the possible role of ET receptor selectivity in the vascular remodelling process in PAH and whether selective ET(A) or nonselective ET(A)/ET(B) blockade offers the greatest potential to improve symptoms and alter the clinical course of the disease.


Subject(s)
Antihypertensive Agents/therapeutic use , Endothelin Receptor Antagonists , Endothelin-1/physiology , Hypertension, Pulmonary/drug therapy , Antihypertensive Agents/pharmacology , Humans , Hypertension, Pulmonary/physiopathology , Randomized Controlled Trials as Topic , Vasodilation/drug effects
18.
Eur Respir J ; 33(4): 785-92, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19129292

ABSTRACT

Pulmonary hypertension (PH) is associated with impaired production of the vasodilator nitric oxide (NO). Riociguat (BAY 63-2521; Bayer Healthcare AG, Wuppertal, Germany) acts directly on soluble guanylate cyclase, stimulating the enzyme and increasing sensitivity to low NO levels. The present study evaluates riociguat safety, tolerability and efficacy in patients with moderate-to-severe PH (pulmonary arterial hypertension, distal chronic thromboembolic PH or PH with mild to moderate interstitial lung disease). The optimal tolerated dose was identified by incremental dosing in four patients with PH; pharmacodynamic and pharmacokinetic parameters were assessed following single-dose administration (2.5 mg or 1 mg) in 10 and five patients with PH, respectively. All subjects (n = 19) were analysed for safety and tolerability. Riociguat had a favourable safety profile at single doses < or =2.5 mg. It significantly improved pulmonary haemodynamic parameters and cardiac index in patients with PH in a dose-dependent manner, to a greater extent than inhaled NO. Although riociguat also had significant systemic effects and showed no pulmonary selectivity, mean systolic blood pressure remained >110 mmHg. The present report is the first to describe the use of riociguat in patients with pulmonary hypertension. The drug was well-tolerated and superior to nitric oxide in efficacy and duration. Riociguat, therefore, has potential as a novel therapy for pulmonary hypertension and warrants further investigation.


Subject(s)
Guanylate Cyclase/biosynthesis , Guanylate Cyclase/physiology , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Analysis of Variance , Area Under Curve , Chromatography, High Pressure Liquid , Female , Hemodynamics/drug effects , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Nitric Oxide Synthase Type II/pharmacology , Oxidation-Reduction , Pulmonary Circulation/physiology , Pyrimidines/pharmacokinetics , Soluble Guanylyl Cyclase , Treatment Outcome
19.
Eur Respir J ; 33(4): 861-70, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19010982

ABSTRACT

Bronchopulmonary dysplasia (BPD) is characterised by impaired alveolarisation, inflammation and aberrant vascular development. Phosphodiesterase (PDE) inhibitors can influence cell proliferation, antagonise inflammation and restore vascular development and homeostasis, suggesting a therapeutic potential in BPD. The aim of the present study was to investigate PDE expression in the lung of hyperoxia-exposed mice, and to assess the viability of PDE4 as a therapeutic target in BPD. Newborn C57BL/6N mice were exposed to normoxia or 85% oxygen for 28 days. Animal growth and dynamic respiratory compliance were reduced in animals exposed to hyperoxia, paralleled by decreased septation, airspace enlargement and increased septal wall thickness. Changes were evident after 14 days and were more pronounced after 28 days of hyperoxic exposure. At the mRNA level, PDE1A and PDE4A were upregulated while PDE5A was downregulated under hyperoxia. Immunoblotting confirmed these trends in PDE4A and PDE5A at the protein expression level. Treatment with cilomilast (PDE4 inhibitor, 5 mg.kg(-1).day(-1)) between days 14 and 28 significantly decreased the mean intra-alveolar distance, septal wall thickness and total airspace area and improved dynamic lung compliance. Pharmacological inhibition of phosphodiesterase improved lung alveolarisation in hyperoxia-induced bronchopulmonary dysplasia, and thus may offer a new therapeutic modality in the clinical management of bronchopulmonary dysplasia.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Hyperoxia/enzymology , Phosphodiesterase Inhibitors/pharmacology , Pulmonary Alveoli/enzymology , Animals , Animals, Newborn , Blotting, Western , Lung Compliance/drug effects , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
20.
Eur Respir J ; 32(6): 1639-51, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19043010

ABSTRACT

Hypoxic pulmonary vasoconstriction (HPV), also known as the von Euler-Liljestrand mechanism, is a physiological response to alveolar hypoxia which distributes pulmonary capillary blood flow to alveolar areas of high oxygen partial pressure. Impairment of this mechanism may result in hypoxaemia. Under conditions of chronic hypoxia generalised vasoconstriction of the pulmonary vasculature in concert with hypoxia-induced vascular remodelling leads to pulmonary hypertension. Although the principle of HPV was recognised decades ago, its exact pathway still remains elusive. Neither the oxygen sensing process nor the exact pathway underlying HPV is fully deciphered yet. The effector pathway is suggested to include L-type calcium channels, nonspecific cation channels and voltage-dependent potassium channels, whereas mitochondria and nicotinamide adenine dinucleotide phosphate oxidases are discussed as oxygen sensors. Reactive oxygen species, redox couples and adenosine monophosphate-activated kinases are under investigation as mediators of hypoxic pulmonary vasoconstriction. Moreover, the role of calcium sensitisation, intracellular calcium stores and direction of change of reactive oxygen species is still under debate. In this context the present article focuses on the basic mechanisms of hypoxic pulmonary vasoconstriction and also outlines differences in current concepts that have been suggested for the regulation of hypoxic pulmonary vasoconstriction.


Subject(s)
Hypoxia , Vasoconstriction , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Capillaries/metabolism , Humans , Models, Biological , NADPH Oxidases/metabolism , Oxidation-Reduction , Oxygen/metabolism , Pressure , Pulmonary Artery/pathology , Pulmonary Circulation/physiology , Reactive Oxygen Species/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...