61 results on '"N. Weissmann"'
Search Results
2. Evidence for the Fucoidan/P-Selectin Axis as a Therapeutic Target in Hypoxia-induced Pulmonary Hypertension.
- Author
-
Novoyatleva T, Kojonazarov B, Owczarek A, Veeroju S, Rai N, Henneke I, Böhm M, Grimminger F, Ghofrani HA, Seeger W, Weissmann N, and Schermuly RT
- Subjects
- Animals, Biomarkers blood, Disease Models, Animal, Fucus chemistry, Humans, Hypertension, Pulmonary physiopathology, Hypoxia physiopathology, Male, Mice, Anticoagulants therapeutic use, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary etiology, Hypoxia complications, Myocytes, Smooth Muscle drug effects, P-Selectin blood, Polysaccharides therapeutic use
- Abstract
Rationale: Pulmonary arterial hypertension (PAH) is characterized by vascular remodeling and excessive proliferation of pulmonary artery smooth muscle cells (PASMCs). Fucoidan, a polysaccharidic ligand of the adhesion molecule P-selectin, exhibits antiproliferative properties. The effects of the fucoidan/P-selectin axis on vascular remodeling and pulmonary hypertension (PH) after hypoxia remain unexplored. Objectives: We aimed to evaluate the therapeutic potential of targeting the fucoidan/P-selectin axis in PH. Methods: Mice with PH induced by chronic hypoxia (35 d) were given either fucoidan (from Fucus vesiculosus ) or anti-P-selectin antibody (Rb40.34) during Days 21-35. Right ventricular (RV) function was determined by echocardiography. Vascular morphometry was assessed by immunohistochemistry. Human and experimental PH lungs and PASMCs were used for assessment of P-selectin expression and function. Measurements and Main Results: Fucoidan attenuated chronic hypoxia-induced PH in mice, reducing pulmonary vascular remodeling and restoring RV function. In vitro , fucoidan inhibited hypoxia and growth factor-stimulated PASMC proliferation and migration. Chronic hypoxia caused an upregulation of P-selectin in the medial layer of the small pulmonary arteries. P-selectin was persistently upregulated in PASMCs of human and hypoxia-induced experimental PH. HIF-1α (hypoxia-inducible factor 1α) directly bound to the P-selectin promoter and transcriptionally activated P-selectin in hypoxia. P-selectin blockage resulted in a marked reduction of PASMC proliferation in vitro . Blockage of P-selectin by administration of anti-P-selectin Rb40.34 antibody and P-selectin-deficient mice improved vascular remodeling and restored RV function. Conclusions: Fucoidan is a potent natural adjuvant that represents a promising therapeutic approach for PH. Our data indicate a previously unrecognized role of P-selectin in the proliferative response of PASMCs associated with PH.
- Published
- 2019
- Full Text
- View/download PDF
3. Pulmonary hypertension in chronic lung disease and hypoxia.
- Author
-
Nathan SD, Barbera JA, Gaine SP, Harari S, Martinez FJ, Olschewski H, Olsson KM, Peacock AJ, Pepke-Zaba J, Provencher S, Weissmann N, and Seeger W
- Subjects
- Animals, Antihypertensive Agents therapeutic use, Chronic Disease, Humans, Hypertension, Pulmonary drug therapy, Lung Diseases, Interstitial drug therapy, Lung Diseases, Interstitial pathology, Pulmonary Disease, Chronic Obstructive drug therapy, Hypertension, Pulmonary epidemiology, Hypoxia complications, Lung Diseases, Interstitial complications, Pulmonary Disease, Chronic Obstructive complications
- Abstract
Pulmonary hypertension (PH) frequently complicates the course of patients with various forms of chronic lung disease (CLD). CLD-associated PH (CLD-PH) is invariably associated with reduced functional ability, impaired quality of life, greater oxygen requirements and an increased risk of mortality. The aetiology of CLD-PH is complex and multifactorial, with differences in the pathogenic sequelae between the diverse forms of CLD. Haemodynamic evaluation of PH severity should be contextualised within the extent of the underlying lung disease, which is best gauged through a combination of physiological and imaging assessment. Who, when, if and how to screen for PH will be addressed in this article, as will the current state of knowledge with regard to the role of treatment with pulmonary vasoactive agents. Although such therapy cannot be endorsed given the current state of findings, future studies in this area are strongly encouraged., Competing Interests: Conflict of interest: S.D. Nathan is a consultant for and has received research funding from Bellerophon, United Therapeutics and Bayer Pharmaceuticals; is a consultant for Third Pole and Actelion, and is a consultant for, has received research funding from and is on the speakers’ bureau of Roche-Genentech and Boehringer Ingelheim. Conflict of interest: J.A. Barbera reports grants and personal fees from Actelion and MSD, personal fees from Arena, and grants from Bayer and GSK, outside the submitted work. Conflict of interest: S.P. Gaine reports personal fees from Actelion, United Therapeutics, MSD and GSK, outside the submitted work. Conflict of interest: S. Harari has received grants for research and speakers fees from Actelion, Boehringer Ingelheim and Roche. Conflict of interest: F.J. Martinez has received grants from the NIH (IPF UO1, COPD UO1/RO1); personal fees, honoraria and non-personal travel support from the American College of Chest Physicians, Continuing Education, ConCert, Inova Fairfax Health System, MD Magazine, Miller Communications, National Association for Continuing Education, Novartis, Pearl Pharmaceuticals, PeerView Communications, Prime Communications, Puerto Rican Respiratory Society, Roche, Sunovion, Theravance, Potomac, University of Alabama Birmingham and Zambon; personal fees and non-personal or non-financial travel support from AstraZeneca; personal fees, non-personal travel support, and non-financial support for data and safety monitoring board work from Boehringer Ingelheim; personal fees, honoraria, non-personal travel support, and non-financial support for data and safety monitoring board work from Genentech and GlaxoSmithKline; personal fees and honoraria from Columbia University, Integritas, Methodist Hospital Brooklyn, New York University, Unity, UpToDate, WebMD/MedScape, Western Connecticut Health Network, Academic CME, Patara, PlatformIQ, American Thoracic Society, Rockpointe and Rare Disease Healthcare Communications; non-personal travel support from Nitto; personal fees, honoraria, travel support and non-personal travel support from Chiesi; personal fees, honoraria and travel support from Physicians Education Resource and Teva; honoraria and travel support from Canadian Respiratory Network; personal fees from France Foundation; and has participated on scientific advisory boards (no direct financial compensation) for ProterrixBio and Bridge Biotherapeutics; participated on IPF study steering committees (no direct financial compensation) for Afferent/Merck, Gilead, Veracyte, Prometic, Bayer and ProMedior; and participated on an IPF study steering committee and data safety monitoring board (no direct financial compensation) for Biogen. Conflict of interest: H. Olschewski reports personal fees and non-financial support from Bayer, MSD, Pfizer and Novartis, grants, personal fees and non-financial support from Actelion, grants from Inventiva, and personal fees from Bellerophon, outside the submitted work; and is part-time employee of the Ludwig Boltzmann Institute for Lung Vascular Research. Conflict of interest: K.M. Olsson received fees for talks and consulting work from Actelion, Bayer, GSK, Pfizer and United Therapeutics. Conflict of interest: A.J. Peacock has received research grants and personal fees from Actelion Pharmaceuticals, Bayer, GSK, MSD, Pfizer and United Therapeutics, outside the submitted work. Conflict of interest: J. Pepke-Zaba is a member of the advisory boards for Actelion, Merck, Bayer and GSK, has received grants, personal fees and non-financial support from Actelion, Merck and Bayer, and personal fees from GSK. Conflict of interest: S. Provencher has received research grants from Actelion Pharmaceuticals and Boehringer Ingelheim, and has received speaker fees from Actelion Pharmaceuticals. Conflict of interest: N. Weissmann has nothing to disclose. Conflict of interest: W. Seeger has received consultancy fees from Bayer AG, United Therapeutics, Liquidia, Vectura and Novartis., (Copyright ©ERS 2019.)
- Published
- 2019
- Full Text
- View/download PDF
4. Hypoxic pulmonary vasoconstriction in isolated mouse pulmonary arterial vessels.
- Author
-
Strielkov I, Krause NC, Sommer N, Schermuly RT, Ghofrani HA, Grimminger F, Gudermann T, Dietrich A, and Weissmann N
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Animals, Endothelium, Vascular physiopathology, Female, In Vitro Techniques, Male, Mice, Mice, Knockout, Muscle Tonus, Muscle, Smooth, Vascular, Myography, Potassium Chloride pharmacology, TRPC Cation Channels genetics, TRPC6 Cation Channel, Vasoconstrictor Agents pharmacology, Hypoxia physiopathology, Pulmonary Artery physiopathology, Pulmonary Circulation, Vasoconstriction
- Abstract
New Findings: What is the central question of this study? Hypoxic pulmonary vasoconstriction has never been characterized in isolated mouse pulmonary arteries of different generations in detail. What is the main finding and its importance? We found that only small intrapulmonary arteries (80-200 μm in diameter) exhibit hypoxic pulmonary vasoconstriction. The observed response was sustained, significantly potentiated by depolarization-induced preconstriction and not dependent on the endothelium or TRPC6 channels., Abstract: Hypoxic pulmonary vasoconstriction (HPV) is a physiological response of pulmonary arteries, which adapts lung perfusion to regional ventilation. The properties of HPV vary significantly between animal species. Despite extensive use of mouse models in studies of HPV, this physiological response has never been characterized in isolated mouse pulmonary arteries in detail. Using wire myography, we investigated the effect of 80 min exposure to hypoxia on the tone in mouse pulmonary arteries of different generations in the presence and absence of preconstriction. Hypoxia induced a sustained relaxation in non-preconstricted extrapulmonary arteries (500-700 μm in diameter), but not in the presence of KCl-induced preconstriction. Large intrapulmonary arteries (450-650 μm in diameter) did not exhibit a significant response to the hypoxic challenge. In contrast, in small intrapulmonary arteries (80-200 μm in diameter), hypoxia elicited a slowly developing sustained constriction, which was independent of the endothelium. The response was significantly potentiated in arteries preconstricted with KCl, but not with U46619. Hypoxic pulmonary vasoconstriction was not altered in pulmonary arteries of TRPC6-deficient mice, which suggests that this response corresponds to the sustained phase of biphasic HPV observed earlier in isolated, buffer-perfused and ventilated mouse lungs. In conclusion, we have established a protocol that allows the study of sustained HPV in isolated mouse pulmonary arteries. The data obtained might be useful for future studies of the mechanisms of HPV in mice., (© 2018 The Authors. Experimental Physiology © 2018 The Physiological Society.)
- Published
- 2018
- Full Text
- View/download PDF
5. Recent advances in oxygen sensing and signal transduction in hypoxic pulmonary vasoconstriction.
- Author
-
Strielkov I, Pak O, Sommer N, and Weissmann N
- Subjects
- Animals, Electron Transport Complex IV metabolism, Humans, Myocytes, Smooth Muscle metabolism, Reactive Oxygen Species metabolism, Hypertension, Pulmonary physiopathology, Hypoxia physiopathology, Oxygen metabolism, Signal Transduction, Vasoconstriction
- Abstract
Hypoxic pulmonary vasoconstriction (HPV) is a physiological reaction, which adapts lung perfusion to regional ventilation and optimizes gas exchange. Impaired HPV may cause systemic hypoxemia, while generalized HPV contributes to the development of pulmonary hypertension. The triggering mechanisms underlying HPV are still not fully elucidated. Several hypotheses are currently under debate, including a possible decrease as well as an increase in reactive oxygen species as a triggering event. Recent findings suggest an increase in the production of reactive oxygen species in pulmonary artery smooth muscle cells by complex III of the mitochondrial electron transport chain and occurrence of oxygen sensing at complex IV. Other essential components are voltage-dependent potassium and possibly L-type, transient receptor potential channel 6, and transient receptor potential vanilloid 4 channels. The release of arachidonic acid metabolites appears also to be involved in HPV regulation. Further investigation of the HPV mechanisms will facilitate the development of novel therapeutic strategies for the treatment of HPV-related disorders.
- Published
- 2017
- Full Text
- View/download PDF
6. TASK-1 potassium channel is not critically involved in mediating hypoxic pulmonary vasoconstriction of murine intra-pulmonary arteries.
- Author
-
Murtaza G, Mermer P, Goldenberg A, Pfeil U, Paddenberg R, Weissmann N, Lochnit G, and Kummer W
- Subjects
- Animals, Arachidonic Acids pharmacology, Endocannabinoids pharmacology, Female, Lung metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Nerve Tissue Proteins genetics, Polyunsaturated Alkamides pharmacology, Potassium Channels, Tandem Pore Domain genetics, Pulmonary Artery drug effects, RNA, Messenger genetics, Hypoxia physiopathology, Nerve Tissue Proteins physiology, Potassium Channels, Tandem Pore Domain physiology, Pulmonary Artery physiopathology, Vasoconstriction physiology
- Abstract
The two-pore domain potassium channel KCNK3 (TASK-1) is expressed in rat and human pulmonary artery smooth muscle cells. There, it is associated with hypoxia-induced signalling, and its dysfunction is linked to pathogenesis of human pulmonary hypertension. We here aimed to determine its role in hypoxic pulmonary vasoconstriction (HPV) in the mouse, and hence the suitability of this model for further mechanistic investigations, using appropriate inhibitors and TASK-1 knockout (KO) mice. RT-PCR revealed expression of TASK-1 mRNA in murine lungs and pre-acinar pulmonary arteries. Protein localization by immunohistochemistry and western blot was unreliable since all antibodies produced labelling also in TASK-1 KO organs/tissues. HPV was investigated by videomorphometric analysis of intra- (inner diameter: 25-40 μm) and pre-acinar pulmonary arteries (inner diameter: 41-60 μm). HPV persisted in TASK-1 KO intra-acinar arteries. Pre-acinar arteries developed initial HPV, but the response faded earlier (after 30 min) in KO vessels. This HPV pattern was grossly mimicked by the TASK-1 inhibitor anandamide in wild-type vessels. Hypoxia-provoked rise in pulmonary arterial pressure (PAP) in isolated ventilated lungs was affected neither by TASK-1 gene deficiency nor by the TASK-1 inhibitor A293. TASK-1 is dispensable for initiating HPV of murine intra-pulmonary arteries, but participates in sustained HPV specifically in pre-acinar arteries. This does not translate into abnormal rise in PAP. While there is compelling evidence that TASK-1 is involved in the pathogenesis of pulmonary arterial hypertension in humans, the mouse does not appear to serve as a suitable model to study the underlying molecular mechanisms.
- Published
- 2017
- Full Text
- View/download PDF
7. Oxygen sensing and signal transduction in hypoxic pulmonary vasoconstriction.
- Author
-
Sommer N, Strielkov I, Pak O, and Weissmann N
- Subjects
- Calcium metabolism, Calcium Channels metabolism, Calcium Channels, L-Type metabolism, Humans, Hypertension, Pulmonary physiopathology, Lung blood supply, Mitochondria metabolism, Muscle Contraction, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, NADPH Oxidases metabolism, Nitric Oxide metabolism, Potassium Channels, Voltage-Gated metabolism, Pulmonary Artery metabolism, Signal Transduction, TRPV Cation Channels metabolism, Ventilation-Perfusion Ratio, Hypertension, Pulmonary metabolism, Hypoxia metabolism, Lung metabolism, Myocytes, Smooth Muscle metabolism, Oxygen metabolism, Pulmonary Circulation physiology, Reactive Oxygen Species metabolism, Vasoconstriction physiology
- Abstract
Hypoxic pulmonary vasoconstriction (HPV), also known as the von Euler-Liljestrand mechanism, is an essential response of the pulmonary vasculature to acute and sustained alveolar hypoxia. During local alveolar hypoxia, HPV matches perfusion to ventilation to maintain optimal arterial oxygenation. In contrast, during global alveolar hypoxia, HPV leads to pulmonary hypertension. The oxygen sensing and signal transduction machinery is located in the pulmonary arterial smooth muscle cells (PASMCs) of the pre-capillary vessels, albeit the physiological response may be modulated in vivo by the endothelium. While factors such as nitric oxide modulate HPV, reactive oxygen species (ROS) have been suggested to act as essential mediators in HPV. ROS may originate from mitochondria and/or NADPH oxidases but the exact oxygen sensing mechanisms, as well as the question of whether increased or decreased ROS cause HPV, are under debate. ROS may induce intracellular calcium increase and subsequent contraction of PASMCs via direct or indirect interactions with protein kinases, phospholipases, sarcoplasmic calcium channels, transient receptor potential channels, voltage-dependent potassium channels and L-type calcium channels, whose relevance may vary under different experimental conditions. Successful identification of factors regulating HPV may allow development of novel therapeutic approaches for conditions of disturbed HPV., (Copyright ©ERS 2016.)
- Published
- 2016
- Full Text
- View/download PDF
8. NADPH oxidases-do they play a role in TRPC regulation under hypoxia?
- Author
-
Malczyk M, Veith C, Schermuly RT, Gudermann T, Dietrich A, Sommer N, Weissmann N, and Pak O
- Subjects
- Animals, Humans, Reactive Oxygen Species, Signal Transduction, Hypoxia metabolism, NADPH Oxidases metabolism, TRPC Cation Channels metabolism
- Abstract
In the lung, acute alveolar hypoxia causes hypoxic pulmonary vasoconstriction (HPV) to maintain ventilation perfusion matching and thus optimal oxygenation of blood. In contrast, global chronic hypoxia triggers a pathological thickening of pulmonary arterial walls, called pulmonary vascular remodelling, leading to persistence of pulmonary hypertension (PH). Moreover, ischaemia or hypoxia can lead to a damage of pulmonary endothelial cells with subsequent oedema formation. Alterations in reactive oxygen species (ROS) have been suggested as a crucial mediator of such responses. Among the various sources of cellular ROS production, NADPH oxidases (NOXs) have been found to contribute to these physiological and pathophysiological signalling processes. NOXs are the only known examples that generate ROS as the primary function of the enzyme system. However, the downstream targets of NOX-derived ROS signalling in hypoxia are still not known. Canonical transient receptor potential (TRPC) channels recently have been recognised as directly or indirectly ROS-activated channels and have been shown to be essential for hypoxia-dependent vascular regulatory processes in the lung. Against this background, we here summarise the current knowledge on NOX-mediated TRPC channel signalling during hypoxia in the pulmonary circulation.
- Published
- 2016
- Full Text
- View/download PDF
9. Pathophysiology and treatment of high-altitude pulmonary vascular disease.
- Author
-
Wilkins MR, Ghofrani HA, Weissmann N, Aldashev A, and Zhao L
- Subjects
- Adaptation, Physiological, Altitude Sickness complications, Altitude Sickness therapy, Calcium metabolism, Chronic Disease, Endothelium, Vascular metabolism, Erythropoietin metabolism, Heart Failure etiology, Heart Failure physiopathology, Hemodynamics, Humans, Hypertension, Pulmonary complications, Hypertension, Pulmonary physiopathology, Hypoxia complications, Hypoxia therapy, Muscle, Smooth metabolism, Pulmonary Edema complications, Vascular Diseases complications, Vascular Diseases therapy, Vascular Remodeling, Vasoconstriction, Altitude Sickness physiopathology, Hypoxia physiopathology, Pulmonary Edema physiopathology, Vascular Diseases physiopathology
- Published
- 2015
- Full Text
- View/download PDF
10. Structural and functional prevention of hypoxia-induced pulmonary hypertension by individualized exercise training in mice.
- Author
-
Weissmann N, Peters DM, Klöpping C, Krüger K, Pilat C, Katta S, Seimetz M, Ghofrani HA, Schermuly RT, Witzenrath M, Seeger W, Grimminger F, and Mooren FC
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases genetics, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Animals, Exercise Therapy, Exercise Tolerance, Gene Expression, Hypertension, Pulmonary etiology, Hypoxia complications, Lung blood supply, Lung metabolism, Male, Mice, Mice, Inbred C57BL, Muscle, Smooth, Vascular physiopathology, Nitric Oxide Synthase genetics, Nitric Oxide Synthase metabolism, Oxygen Consumption, Phosphodiesterase 5 Inhibitors pharmacology, Physical Conditioning, Animal, Piperazines pharmacology, Purines pharmacology, Signal Transduction, Sildenafil Citrate, Sulfones pharmacology, Ventricular Pressure, Hypertension, Pulmonary prevention & control, Hypoxia therapy
- Abstract
Pulmonary hypertension (PH) is a disease with a poor prognosis characterized by a vascular remodeling process and an increase in pulmonary vascular resistance. While a variety of reports demonstrated that exercise training exerts beneficial effects on exercise performance and quality of life in PH patients, it is not known how physical exercise affects vascular remodeling processes occurring in hypoxia-induced PH. Therefore, we investigated the effect of individualized exercise training on the development of hypoxia-induced PH in mice. Training effects were compared with pharmacological treatment with the phosphodiesterase 5 inhibitor Sildenafil or a combination of training plus Sildenafil. Trained mice who received Sildenafil showed a significantly improved walking distance (from 88.9 ± 8.1 to 146.4 ± 13.1 m) and maximum oxygen consumption (from 93.3 ± 2.9 to 105.5 ± 2.2% in combination with Sildenafil, to 102.2 ± 3.0% with placebo) compared with sedentary controls. Right ventricular systolic pressure, measured by telemetry, was at the level of healthy normoxic animals, whereas right heart hypertrophy did not benefit from training. Most interestingly, the increase in small pulmonary vessel muscularization was prevented by training. Respective counterregulatory processes were detected for the nitric oxide-soluble guanylate cyclase-phosphodiesterase system. We conclude that individualized daily exercise can prevent vascular remodeling in hypoxia-induced PH., (Copyright © 2014 the American Physiological Society.)
- Published
- 2014
- Full Text
- View/download PDF
11. Classical transient receptor potential channel 1 in hypoxia-induced pulmonary hypertension.
- Author
-
Malczyk M, Veith C, Fuchs B, Hofmann K, Storch U, Schermuly RT, Witzenrath M, Ahlbrecht K, Fecher-Trost C, Flockerzi V, Ghofrani HA, Grimminger F, Seeger W, Gudermann T, Dietrich A, and Weissmann N
- Subjects
- Animals, Biomarkers metabolism, Blotting, Western, Cells, Cultured, Chronic Disease, Female, Hypertension, Pulmonary etiology, Hypertension, Pulmonary pathology, Lung metabolism, Lung pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Pulmonary Artery metabolism, Pulmonary Artery pathology, Real-Time Polymerase Chain Reaction, TRPC Cation Channels deficiency, Up-Regulation, Hypertension, Pulmonary metabolism, Hypoxia complications, TRPC Cation Channels metabolism
- Abstract
Rationale: Pulmonary hypertension (PH) is a life-threatening disease, characterized by pulmonary vascular remodeling. Abnormal smooth muscle cell proliferation is a primary hallmark of chronic hypoxia-induced PH. Essential for cell growth are alterations in the intracellular Ca(2+) homeostasis. Classical transient receptor potential (TRPC) proteins have been suggested to contribute to PH development, as TRPC1 and TRPC6 are predominantly expressed in precapillary pulmonary arterial smooth muscle cells (PASMC). Studies in a TRPC6-deficient mouse model revealed an essential function of TRPC6 in acute but not in chronic hypoxia., Objectives: We aimed to identify the importance of TRPC1 in the pathogenesis of chronic hypoxia-induced PH in mice., Methods: TRPC1 expression analysis was performed using real-time polymerase chain reaction. TRPC1 function was assessed by in vivo experiments in TRPC1(-/-) animals as well as in isolated precapillary murine PASMC after TRPC1 knockdown by TRPC1-specific small interfering RNAs., Measurements and Main Results: Only TRPC1 mRNA was up-regulated under hypoxia in isolated murine PASMC (1% O2 for 72 h). Hypoxia-induced proliferation of murine PASMC was attenuated in cells treated with small interfering RNA against TRPC1 and in cells isolated from TRPC1(-/-) animals compared with untreated and wild-type cells. TRPC1(-/-) mice did not develop PH in response to chronic hypoxia (FI(O2) 0.10 for 21 d) and had less vascular muscularization but a similar degree of right ventricular hypertrophy compared with wild-type mice., Conclusions: Our results indicate an important role of TRPC1 in pulmonary vascular remodeling underlying the development of hypoxia-induced PH.
- Published
- 2013
- Full Text
- View/download PDF
12. Effects of dimethylarginine dimethylaminohydrolase-1 overexpression on the response of the pulmonary vasculature to hypoxia.
- Author
-
Bakr A, Pak O, Taye A, Hamada F, Hemeida R, Janssen W, Gierhardt M, Ghofrani HA, Seeger W, Grimminger F, Schermuly RT, Witzenrath M, Brandes RP, Huang N, Cooke JP, Weissmann N, and Sommer N
- Subjects
- Amidohydrolases metabolism, Animals, Arginine analogs & derivatives, Blood Vessels drug effects, Blood Vessels physiopathology, Cyclic GMP metabolism, Gene Expression, Hemodynamics drug effects, Hypertension, Pulmonary etiology, Hypertension, Pulmonary metabolism, Hypertension, Pulmonary physiopathology, Hypoxia complications, Hypoxia metabolism, Hypoxia physiopathology, Lung drug effects, Lung physiopathology, Mice, Nitroarginine pharmacology, Organ Culture Techniques, Oxadiazoles pharmacology, Signal Transduction, Vasoconstriction drug effects, Amidohydrolases genetics, Arginine metabolism, Hypertension, Pulmonary genetics, Hypoxia genetics, Lung metabolism, Nitric Oxide metabolism
- Abstract
Acute and sustained hypoxic pulmonary vasoconstriction (HPV), as well as chronic pulmonary hypertension (PH), is modulated by nitric oxide (NO). NO synthesis can be decreased by asymmetric dimethylarginine (ADMA), which is degraded by dimethylarginine dimethylaminohydrolase-1 (DDAH1). We investigated the effects of DDAH1 overexpression (DDAH1(tg)) on HPV and chronic hypoxia-induced PH. HPV was measured during acute (10 min) and sustained (3 h) hypoxia in isolated mouse lungs. Chronic PH was induced by the exposure of mice to 4 weeks of hypoxia. ADMA and cyclic 3',5'-guanosine monophosphate (cGMP) were determined by ELISA, and NO generation was determined by chemiluminescence. DDAH1 overexpression exerted no effects on acute HPV. However, DDAH1(tg) mice showed decreased sustained HPV compared with wild-type (WT) mice. Concomitantly, ADMA was decreased, and concentrations of NO and cGMP were significantly increased in DDAH1(tg). The administration of either Nω-nitro-l-arginine or 1H-[1,2,4]oxadiazolo [4,3-a]quinoxalin-1-one potentiated sustained HPV and partly abolished the differences in sustained HPV between WT and DDAH1(tg) mice. The overexpression of DDAH1 exerted no effect on the development of chronic hypoxia-induced PH. DDAH1 overexpression selectively decreased the sustained phase of HPV, partly via activation of the NO-cGMP pathway. Thus, increased ADMA concentrations modulate sustained HPV, but not acute HPV or chronic hypoxia-induced PH.
- Published
- 2013
- Full Text
- View/download PDF
13. Mitochondrial complex II is essential for hypoxia-induced pulmonary vasoconstriction of intra- but not of pre-acinar arteries.
- Author
-
Paddenberg R, Tiefenbach M, Faulhammer P, Goldenberg A, Gries B, Pfeil U, Lips KS, Piruat JI, López-Barneo J, Schermuly RT, Weissmann N, and Kummer W
- Subjects
- Animals, Blood Pressure physiology, Electron Transport Complex II genetics, Electron Transport Complex II physiology, Heterozygote, Lung metabolism, Membrane Proteins genetics, Membrane Proteins physiology, Mice, Mice, Mutant Strains, Models, Animal, RNA, Messenger metabolism, Succinate Dehydrogenase genetics, Hypoxia physiopathology, Lung blood supply, Pulmonary Artery physiopathology, Succinate Dehydrogenase physiology, Vasoconstriction physiology
- Abstract
Aims: Alveolar hypoxia acutely elicits contraction of pulmonary arteries, leading to a rise in pulmonary arterial pressure (PAP) and shifting blood to better ventilated areas of the lung. The molecular mechanisms underlying this hypoxic pulmonary vasoconstriction (HPV) are still incompletely understood. Here, we investigated the role of succinate dehydrogenase (SDH; synonymous to mitochondrial complex II) in HPV, with particular emphasis on regional differences along the vascular bed and consequences for PAP and perfusion-to-ventilation matching, using mutant mice heterozygous for the SDHD subunit of complex II (SDHD(+/-))., Methods and Results: Western blots revealed reduced protein content of complex II subunits SDHA, SDHB, and SDHC in lungs of SDHD(+/-) mice, despite unaffected mRNA content as determined by real-time PCR. Hypoxic pulmonary vasoconstriction of small (20-50 µm) intra-acinar and larger (51-100 µm) pre-acinar arteries was evaluated by videomorphometric analysis of precision-cut lung slices. The hypoxic response was detectable in pre-acinar arteries but absent from intra-acinar arteries of SDHD(+/-) mice. In isolated perfused lungs, basal PAP and its hypoxia-induced increase were indistinguishable between both mouse strains. Arterial oxygenation was measured after provocation of regional ventilatory failure by tracheal fluid instillation in anaesthetized mice, and it declined more in SDHD(+/-) than in wild-type mice., Conclusion: SDHD is required for the formation of a stable mitochondrial complex II and it is selectively important for HPV of intra-acinar vessels. This specialized vascular segment participates in perfusion-to-ventilation matching but does not significantly contribute to the acute hypoxic rise in PAP that results from more proximal vasoconstriction.
- Published
- 2012
- Full Text
- View/download PDF
14. Hypoxia induces Kv channel current inhibition by increased NADPH oxidase-derived reactive oxygen species.
- Author
-
Mittal M, Gu XQ, Pak O, Pamenter ME, Haag D, Fuchs DB, Schermuly RT, Ghofrani HA, Brandes RP, Seeger W, Grimminger F, Haddad GG, and Weissmann N
- Subjects
- Acetophenones pharmacology, Animals, Cells, Cultured, Hypertension, Pulmonary etiology, Hypoxia complications, Male, Mice, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle pathology, NADPH Oxidase 4, NADPH Oxidases genetics, Oxidation-Reduction drug effects, Protein Transport, Pulmonary Artery pathology, RNA, Small Interfering genetics, Rats, Rats, Sprague-Dawley, Reactive Oxygen Species metabolism, Hypertension, Pulmonary metabolism, Hypoxia metabolism, Kv1.5 Potassium Channel metabolism, Myocytes, Smooth Muscle metabolism, NADPH Oxidases metabolism
- Abstract
There is current discussion whether reactive oxygen species are up- or downregulated in the pulmonary circulation during hypoxia, from which sources (i.e., mitochondria or NADPH oxidases) they are derived, and what the downstream targets of ROS are. We recently showed that the NADPH oxidase homolog NOX4 is upregulated in hypoxia-induced pulmonary hypertension in mice and contributes to the vascular remodeling in pulmonary hypertension. We here tested the hypothesis that NOX4 regulates K(v) channels via an increased ROS formation after prolonged hypoxia. We showed that (1) NOX4 is upregulated in hypoxia-induced pulmonary hypertension in rats and isolated rat pulmonary arterial smooth muscle cells (PASMC) after 3days of hypoxia, and (2) that NOX4 is a major contributor to increased reactive oxygen species (ROS) after hypoxia. Our data indicate colocalization of K(v)1.5 and NOX4 in isolated PASMC. The NADPH oxidase inhibitor and ROS scavenger apocynin as well as NOX4 siRNA reversed the hypoxia-induced decrease in K(v) current density whereas the protein levels of the channels remain unaffected by siNOX4 treatment. Determination of cysteine oxidation revealed increased NOX4-mediated K(v)1.5 channel oxidation. We conclude that sustained hypoxia decreases K(v) channel currents by a direct effect of a NOX4-derived increase in ROS., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
15. Effects of hypercapnia and NO synthase inhibition in sustained hypoxic pulmonary vasoconstriction.
- Author
-
Ketabchi F, Ghofrani HA, Schermuly RT, Seeger W, Grimminger F, Egemnazarov B, Shid-Moosavi SM, Dehghani GA, Weissmann N, and Sommer N
- Subjects
- Acidosis drug therapy, Acidosis physiopathology, Animals, Enzyme Inhibitors pharmacology, Hypercapnia drug therapy, Hypoxia drug therapy, Imines pharmacology, Lung blood supply, Lung drug effects, Male, Nitric Oxide Synthase Type II antagonists & inhibitors, Nitric Oxide Synthase Type III antagonists & inhibitors, Nitroarginine pharmacology, Pulmonary Circulation drug effects, Pulmonary Circulation physiology, Rabbits, Sodium Bicarbonate pharmacology, Vasoconstriction drug effects, Hypercapnia physiopathology, Hypoxia physiopathology, Lung physiopathology, Nitric Oxide Synthase Type II physiology, Nitric Oxide Synthase Type III physiology, Vasoconstriction physiology
- Abstract
Background: Acute respiratory disorders may lead to sustained alveolar hypoxia with hypercapnia resulting in impaired pulmonary gas exchange. Hypoxic pulmonary vasoconstriction (HPV) optimizes gas exchange during local acute (0-30 min), as well as sustained (> 30 min) hypoxia by matching blood perfusion to alveolar ventilation. Hypercapnia with acidosis improves pulmonary gas exchange in repetitive conditions of acute hypoxia by potentiating HPV and preventing pulmonary endothelial dysfunction. This study investigated, if the beneficial effects of hypercapnia with acidosis are preserved during sustained hypoxia as it occurs, e.g in permissive hypercapnic ventilation in intensive care units. Furthermore, the effects of NO synthase inhibitors under such conditions were examined., Method: We employed isolated perfused and ventilated rabbit lungs to determine the influence of hypercapnia with or without acidosis (pH corrected with sodium bicarbonate), and inhibitors of endothelial as well as inducible NO synthase on acute or sustained HPV (180 min) and endothelial permeability., Results: In hypercapnic acidosis, HPV was intensified in sustained hypoxia, in contrast to hypercapnia without acidosis when HPV was amplified during both phases. L-NG-Nitroarginine (L-NNA), a non-selective NO synthase inhibitor, enhanced acute as well as sustained HPV under all conditions, however, the amplification of sustained HPV induced by hypercapnia with or without acidosis compared to normocapnia disappeared. In contrast 1400 W, a selective inhibitor of inducible NO synthase (iNOS), decreased HPV in normocapnia and hypercapnia without acidosis at late time points of sustained HPV and selectively reversed the amplification of sustained HPV during hypercapnia without acidosis. Hypoxic hypercapnia without acidosis increased capillary filtration coefficient (Kfc). This increase disappeared after administration of 1400 W., Conclusion: Hypercapnia with and without acidosis increased HPV during conditions of sustained hypoxia. The increase of sustained HPV and endothelial permeability in hypoxic hypercapnia without acidosis was iNOS dependent.
- Published
- 2012
- Full Text
- View/download PDF
16. Hypoxia enhances platelet-derived growth factor signaling in the pulmonary vasculature by down-regulation of protein tyrosine phosphatases.
- Author
-
ten Freyhaus H, Dagnell M, Leuchs M, Vantler M, Berghausen EM, Caglayan E, Weissmann N, Dahal BK, Schermuly RT, Ostman A, Kappert K, and Rosenkranz S
- Subjects
- Animals, Cell Proliferation, Cells, Cultured, Chemotaxis physiology, Down-Regulation physiology, Humans, Hypertension, Pulmonary etiology, Hypertension, Pulmonary physiopathology, Lung blood supply, Mice, Mice, Inbred C57BL, Muscle, Smooth, Vascular physiopathology, Receptor, Platelet-Derived Growth Factor beta physiology, Signal Transduction physiology, Hypoxia physiopathology, Platelet-Derived Growth Factor physiology, Protein Tyrosine Phosphatases physiology, Pulmonary Artery physiopathology
- Abstract
Rationale: Platelet-derived growth factor (PDGF) plays a pivotal role in the pathobiology of pulmonary hypertension (PH) because it promotes pulmonary vascular remodeling. PH is frequently associated with pulmonary hypoxia., Objectives: To investigate whether hypoxia alters PDGF β receptor (βPDGFR) signaling in the pulmonary vasculature., Methods: The impact of chronic hypoxia on signal transduction by the βPDGFR was measured in human pulmonary arterial smooth muscle cells (hPASMC) in vitro, and in mice with hypoxia-induced PH in vivo., Measurements and Main Results: Chronic hypoxia significantly enhanced PDGF-BB-dependent proliferation and chemotaxis of hPASMC. Pharmacologic inhibition of PI3 kinase (PI3K) and PLCγ abrogated these events under both normoxia and hypoxia. Although hypoxia did not affect βPDGFR expression, it increased the ligand-induced tyrosine phosphorylation of the receptor, particularly at binding sites for PI3K (Y751) and PLCγ (Y1021). The activated βPDGFR is dephosphorylated by protein tyrosine phosphatases (PTPs). Interestingly, hypoxia decreased expression of numerous PTPs (T cell PTP, density-enhanced phosphatase-1, PTP1B, and SH2 domain-containing phosphatase-2), resulting in reduced PTP activity. Hypoxia-inducible factor (HIF)-1α is involved in this regulation of gene expression, because hypoxia-induced βPDGFR hyperphosphorylation and PTP down-regulation were abolished by HIF-1α siRNA and by the HIF-1α inhibitor 2-methoxyestradiol. βPDGFR hyperphosphorylation and PTP down-regulation were also present in vivo in mice with chronic hypoxia-induced PH., Conclusions: Hypoxia reduces expression and activity of βPDGFR-antagonizing PTPs in a HIF-1α-dependent manner, thereby enhancing receptor activation and proliferation and chemotaxis of hPASMC. Because hyperphosphorylation of the βPDGFR and down-regulation of PTPs occur in vivo, this mechanism likely has significant impact on the development and progression of PH and other hypoxia-associated diseases.
- Published
- 2011
- Full Text
- View/download PDF
17. Diacylglycerol regulates acute hypoxic pulmonary vasoconstriction via TRPC6.
- Author
-
Fuchs B, Rupp M, Ghofrani HA, Schermuly RT, Seeger W, Grimminger F, Gudermann T, Dietrich A, and Weissmann N
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Animals, Diacylglycerol Kinase antagonists & inhibitors, Diacylglycerol Kinase metabolism, Dose-Response Relationship, Drug, Enzyme Inhibitors pharmacology, Estrenes pharmacology, Hypoxia genetics, Hypoxia physiopathology, In Vitro Techniques, Mice, Mice, Inbred C57BL, Mice, Knockout, Perfusion, Piperidines pharmacology, Pyrrolidinones pharmacology, Quinazolinones pharmacology, Signal Transduction, TRPC Cation Channels deficiency, TRPC Cation Channels genetics, TRPC6 Cation Channel, Time Factors, Type C Phospholipases antagonists & inhibitors, Type C Phospholipases metabolism, Vasoconstrictor Agents pharmacology, Diglycerides metabolism, Hypoxia metabolism, Lung blood supply, Pulmonary Circulation drug effects, TRPC Cation Channels metabolism, Vasoconstriction drug effects
- Abstract
Background: Hypoxic pulmonary vasoconstriction (HPV) is an essential mechanism of the lung that matches blood perfusion to alveolar ventilation to optimize gas exchange. Recently we have demonstrated that acute but not sustained HPV is critically dependent on the classical transient receptor potential 6 (TRPC6) channel. However, the mechanism of TRPC6 activation during acute HPV remains elusive. We hypothesize that a diacylglycerol (DAG)-dependent activation of TRPC6 regulates acute HPV., Methods: We investigated the effect of the DAG analog 1-oleoyl-2-acetyl-sn-glycerol (OAG) on normoxic vascular tone in isolated perfused and ventilated mouse lungs from TRPC6-deficient and wild-type mice. Moreover, the effects of OAG, the DAG kinase inhibitor R59949 and the phospholipase C inhibitor U73122 on the strength of HPV were investigated compared to those on non-hypoxia-induced vasoconstriction elicited by the thromboxane mimeticum U46619., Results: OAG increased normoxic vascular tone in lungs from wild-type mice, but not in lungs from TRPC6-deficient mice. Under conditions of repetitive hypoxic ventilation, OAG as well as R59949 dose-dependently attenuated the strength of acute HPV whereas U46619-induced vasoconstrictions were not reduced. Like OAG, R59949 mimicked HPV, since it induced a dose-dependent vasoconstriction during normoxic ventilation. In contrast, U73122, a blocker of DAG synthesis, inhibited acute HPV whereas U73343, the inactive form of U73122, had no effect on HPV., Conclusion: These findings support the conclusion that the TRPC6-dependency of acute HPV is induced via DAG.
- Published
- 2011
- Full Text
- View/download PDF
18. Mitochondrial complex II participates in normoxic and hypoxic regulation of α-keto acids in the murine heart.
- Author
-
Mühling J, Tiefenbach M, López-Barneo J, Piruat JI, García-Flores P, Pfeil U, Gries B, Mühlfeld C, Weigand MA, Kummer W, Weissmann N, and Paddenberg R
- Subjects
- Animals, Blood Pressure physiology, Cardiomegaly complications, Cardiomegaly enzymology, Cardiomegaly pathology, Cardiomegaly physiopathology, Down-Regulation, Heart Ventricles enzymology, Heart Ventricles pathology, Heart Ventricles physiopathology, Heterozygote, Hypoxia complications, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Lung blood supply, Lung physiopathology, Mice, Mitochondria pathology, Mitochondria ultrastructure, Mutation genetics, Myocytes, Cardiac enzymology, Myocytes, Cardiac pathology, Myocytes, Cardiac ultrastructure, Organ Specificity, Protein Stability, Protein Subunits, Succinate Dehydrogenase metabolism, Electron Transport Complex II metabolism, Hypoxia enzymology, Keto Acids metabolism, Mitochondria enzymology, Myocardium enzymology, Myocardium pathology
- Abstract
α-Keto acids (α-KAs) are not just metabolic intermediates but are also powerful modulators of different cellular pathways. Here, we tested the hypothesis that α-KA concentrations are regulated by complex II (succinate dehydrogenase=SDH), which represents an intersection between the mitochondrial respiratory chain for which an important function in cardiopulmonary oxygen sensing has been demonstrated, and the Krebs cycle, a central element of α-KA metabolism. SDH subunit D heterozygous (SDHD(+/-)) and wild-type (WT) mice were housed at normoxia or hypoxia (10% O(2)) for 4 days or 3 weeks, and right ventricular pressure, right ventricle/(left ventricle+septum) ratio, cardiomyocyte ultrastructure, pulmonary vascular remodelling, ventricular complex II subunit expression, SDH activity and α-KA concentrations were analysed. In both strains, hypoxia induced increases in right ventricular pressure and enhanced muscularization of distal pulmonary arteries. Right ventricular hypertrophy was less severe in SDHD(+/-) mice although the cardiomyocyte ultrastructure and mitochondrial morphometric parameters were unchanged. Protein amounts of SDHA, SDHB and SDHC, and SDH activity were distinctly reduced in SDHD(+/-) mice. In normoxic SDHD(+/-) mice, α-ketoisocaproate concentration was lowered to 50% as compared to WT animals. Right/left ventricular concentration differences and the hypoxia-induced decline in individual α-KAs were less pronounced in SDHD(+/-) animals indicating that mitochondrial complex II participates in the adjustment of cardiac α-KA concentrations both under normoxic and hypoxic conditions. These characteristics are not related to the hemodynamic consequences of hypoxia-induced pulmonary vascular remodelling, since its extent and right ventricular pressure were not affected in SDHD(+/-) mice albeit right ventricular hypertrophy was attenuated., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
19. Mitochondrial cytochrome redox states and respiration in acute pulmonary oxygen sensing.
- Author
-
Sommer N, Pak O, Schörner S, Derfuss T, Krug A, Gnaiger E, Ghofrani HA, Schermuly RT, Huckstorf C, Seeger W, Grimminger F, and Weissmann N
- Subjects
- 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, Cytochromes metabolism, Hypoxia metabolism, Lung metabolism, Mitochondria metabolism, Muscle, Smooth, Vascular metabolism, Oxygen Consumption physiology
- 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.
- Published
- 2010
- Full Text
- View/download PDF
20. Nebulization of the acidified sodium nitrite formulation attenuates acute hypoxic pulmonary vasoconstriction.
- Author
-
Egemnazarov B, Schermuly RT, Dahal BK, Elliott GT, Hoglen NC, Surber MW, Weissmann N, Grimminger F, Seeger W, and Ghofrani HA
- Subjects
- Acute Disease, Administration, Inhalation, Animals, Blood Pressure drug effects, Cardiac Output drug effects, Chemistry, Pharmaceutical, Disease Models, Animal, Dose-Response Relationship, Drug, Exhalation, Hydrogen-Ion Concentration, Hypoxia physiopathology, Male, Nebulizers and Vaporizers, Nitrates blood, Nitric Oxide metabolism, Perfusion, Pulmonary Artery physiopathology, Rabbits, Time Factors, Hypoxia drug therapy, Pulmonary Artery drug effects, Sodium Nitrite administration & dosage, Vasoconstriction drug effects, Vasodilator Agents administration & dosage
- Abstract
Background: Generalized hypoxic pulmonary vasoconstriction (HPV) occurring during exposure to hypoxia is a detrimental process resulting in an increase in lung vascular resistance. Nebulization of sodium nitrite has been shown to inhibit HPV. The aim of this project was to investigate and compare the effects of nebulization of nitrite and different formulations of acidified sodium nitrite on acute HPV., Methods: Ex vivo isolated rabbit lungs perfused with erythrocytes in Krebs-Henseleit buffer (adjusted to 10% hematocrit) and in vivo anesthetized catheterized rabbits were challenged with periods of hypoxic ventilation alternating with periods of normoxic ventilation. After baseline hypoxic challenges, vehicle, sodium nitrite or acidified sodium nitrite was delivered via nebulization. In the ex vivo model, pulmonary arterial pressure and nitric oxide concentrations in exhaled gas were monitored. Nitrite and nitrite/nitrate were measured in samples of perfusion buffer. Pulmonary arterial pressure, systemic arterial pressure, cardiac output and blood gases were monitored in the in vivo model., Results: In the ex vivo model, nitrite nebulization attenuated HPV and increased nitric oxide concentrations in exhaled gas and nitrite concentrations in the perfusate. The acidified forms of sodium nitrite induced higher levels of nitric oxide in exhaled gas and had longer vasodilating effects compared to nitrite alone. All nitrite formulations increased concentrations of circulating nitrite to the same degree. In the in vivo model, inhaled nitrite inhibited HPV, while pulmonary arterial pressure, cardiac output and blood gases were not affected. All nitrite formulations had similar potency to inhibit HPV. The tested concentration of appeared tolerable., Conclusion: Nitrite alone and in acidified forms effectively and similarly attenuates HPV. However, acidified nitrite formulations induce a more pronounced increase in nitric oxide exhalation.
- Published
- 2010
- Full Text
- View/download PDF
21. Hypoxia-induced pulmonary hypertension: comparison of soluble epoxide hydrolase deletion vs. inhibition.
- Author
-
Keserü B, Barbosa-Sicard E, Schermuly RT, Tanaka H, Hammock BD, Weissmann N, Fisslthaler B, and Fleming I
- Subjects
- Animals, Cardiomegaly etiology, Epoxide Hydrolases antagonists & inhibitors, Epoxide Hydrolases genetics, Humans, Hypertension, Pulmonary enzymology, Hypoxia enzymology, Hypoxia physiopathology, Mice, Mice, Inbred C57BL, Physical Conditioning, Animal, Promoter Regions, Genetic, Vasoconstriction, Epoxide Hydrolases physiology, Hypertension, Pulmonary etiology, Hypoxia complications
- Abstract
Aims: The C-terminal domain of the soluble epoxide hydrolase (sEH) metabolizes epoxyeicosatrienoic acids (EETs) to their less active diols, while the N-terminal domain demonstrates lipid phosphatase activity. As EETs are potent vasoconstrictors in the pulmonary circulation, we assessed the development of pulmonary hypertension induced by exposure to hypoxia (10% O(2)) for 21 days in wild-type (WT) and sEH(-/-) mice and compared the effects with chronic (4 months) sEH inhibition., Methods and Results: In isolated lungs from WT mice, acute hypoxic vasoconstriction (HPV) was potentiated by sEH inhibition and attenuated by an EET antagonist. After prolonged hypoxia, the acute HPV and sensitivity to the EET antagonist were increased, but potentiation of vasoconstriction following sEH inhibition was not evident. Chronic hypoxia also stimulated the muscularization of pulmonary arteries and decreased sEH expression in WT mice. In normoxic sEH(-/-) mice, acute HPV and small artery muscularization were greater than that in WT lungs and enhanced muscularization was accompanied with decreased voluntary exercise capacity. Acute HPV in sEH(-/-) mice was insensitive to sEH inhibition but inhibited by the EET antagonist and chronic hypoxia induced an exaggerated pulmonary vascular remodelling. In WT mice, chronic sEH inhibition increased serum EET levels but failed to affect acute HPV, right ventricle weight, pulmonary artery muscularization, or voluntary running distance. In human donor lungs, the sEH was expressed in the wall of pulmonary arteries, however, sEH expression was absent in samples from patients with pulmonary hypertension., Conclusion: These data suggest that a decrease in sEH expression is intimately linked to pathophysiology of hypoxia-induced pulmonary remodelling and hypertension. However, as sEH inhibitors do not promote the development of pulmonary hypertension it seems likely that the N-terminal lipid phosphatase may play a role in the development of this disease.
- Published
- 2010
- Full Text
- View/download PDF
22. The role of classical transient receptor potential channels in the regulation of hypoxic pulmonary vasoconstriction.
- Author
-
Fuchs B, Dietrich A, Gudermann T, Kalwa H, Grimminger F, and Weissmann N
- Subjects
- Animals, Calcium metabolism, Humans, Phylogeny, Protein Isoforms chemistry, Protein Isoforms classification, Protein Isoforms genetics, Protein Structure, Secondary, Signal Transduction physiology, Transient Receptor Potential Channels chemistry, Transient Receptor Potential Channels classification, Transient Receptor Potential Channels genetics, Hypoxia metabolism, Lung blood supply, Lung metabolism, Protein Isoforms metabolism, Pulmonary Artery physiology, Transient Receptor Potential Channels metabolism, Vasoconstriction physiology
- Abstract
Hypoxic pulmonary vasoconstriction (HPV) is an essential mechanism of the lung matching blood perfusion to ventilation during local alveolar hypoxia. HPV thus optimizes pulmonary gas exchange. In contrast chronic and generalized hypoxia leads to pulmonary vascular remodeling with subsequent pulmonary hypertension and right heart hypertrophy. Among other non-selective cation channels, the family of classical transient receptor potential channels (TRPC) has been shown to be expressed in pulmonary arterial smooth muscle cells. Among this family, TRPC6 is essential for the regulation of acute HPV in mice. Against this background, in this chapter we give an overview about the TRPC family and their role in HPV.
- Published
- 2010
- Full Text
- View/download PDF
23. Effects of hypercapnia with and without acidosis on hypoxic pulmonary vasoconstriction.
- Author
-
Ketabchi F, Egemnazarov B, Schermuly RT, Ghofrani HA, Seeger W, Grimminger F, Shid-Moosavi M, Dehghani GA, Weissmann N, and Sommer N
- Subjects
- Acidosis, Respiratory physiopathology, Animals, Capillary Permeability drug effects, Endothelium, Vascular drug effects, Endothelium, Vascular physiopathology, Hydrogen-Ion Concentration drug effects, In Vitro Techniques, Lung drug effects, Lung physiopathology, Male, Metalloporphyrins pharmacology, Nitric Oxide metabolism, Nitroarginine pharmacology, Partial Pressure, Pulmonary Ventilation drug effects, Rabbits, Vasoconstriction drug effects, Acidosis, Respiratory complications, Hypercapnia complications, Hypercapnia physiopathology, Hypoxia complications, Hypoxia physiopathology, Lung blood supply, Vasoconstriction physiology
- Abstract
Acute respiratory disorders and permissive hypercapnic strategy may lead to alveolar hypoxia and hypercapnic acidosis. However, the effects of hypercapnia with or without acidosis on hypoxic pulmonary vasoconstriction (HPV) and oxygen diffusion capacity of the lung are controversial. We investigated the effects of hypercapnic acidosis and hypercapnia with normal pH (pH corrected with sodium bicarbonate) on HPV, capillary permeability, gas exchange, and ventilation-perfusion matching in the isolated ventilated-perfused rabbit lung. No alteration in vascular tone was noted during normoxic hypercapnia with or without acidosis compared with normoxic normocapnia. Hypercapnia with normal pH resulted in a transient increase in HPV during the course of consecutive ventilation maneuvers, whereas hypercapnic acidosis increased HPV over time. Hypercapnic acidosis decreased exhaled NO during hypoxia more than hypercapnia with normal pH and normocapnia, whereas intravascular NO release was unchanged. However, inhibition of NO synthesis by nitro-L-arginine (L-NNA) resulted in a loss of the increased HPV caused by hypercapnic acidosis but not that caused by hypercapnia with normal pH. Furthermore, capillary permeability increased during hypoxic hypercapnia with normal pH but not hypoxic hypercapnic acidosis. This effect was NO-dependent because it disappeared during L-NNA administration. Ventilation-perfusion matching and arterial PO2 were improved according to the strength of HPV in hypercapnia compared with normocapnia during Tween nebulization-induced lung injury. In conclusion, the increased HPV during hypercapnic acidosis is beneficial to lung gas exchange by improving ventilation-perfusion matching and preserving the capillary barrier function. These effects seem to be linked to NO-mediated pathways.
- Published
- 2009
- Full Text
- View/download PDF
24. The soluble guanylate cyclase activator HMR1766 reverses hypoxia-induced experimental pulmonary hypertension in mice.
- Author
-
Weissmann N, Hackemack S, Dahal BK, Pullamsetti SS, Savai R, Mittal M, Fuchs B, Medebach T, Dumitrascu R, Eickels Mv, Ghofrani HA, Seeger W, Grimminger F, and Schermuly RT
- Subjects
- Animals, Cardiomegaly, Cyclic GMP metabolism, Hemodynamics drug effects, Hypertension, Pulmonary enzymology, Mice, Myocytes, Smooth Muscle drug effects, Myocytes, Smooth Muscle metabolism, Pulmonary Artery cytology, Pulmonary Artery drug effects, Pulmonary Artery metabolism, Soluble Guanylyl Cyclase, Superoxides metabolism, Vasoconstriction drug effects, Guanylate Cyclase metabolism, Hypertension, Pulmonary prevention & control, Hypoxia prevention & control, Receptors, Cytoplasmic and Nuclear metabolism, Sulfonamides pharmacology, ortho-Aminobenzoates pharmacology
- Abstract
Severe pulmonary hypertension (PH) is a disabling disease with high mortality, characterized by pulmonary vascular remodeling and right heart hypertrophy. In mice with PH induced by chronic hypoxia, we examined the acute and chronic effects of the soluble guanylate cyclase (sGC) activator HMR1766 on hemodynamics and pulmonary vascular remodeling. In isolated perfused mouse lungs from control animals, HMR1766 dose-dependently inhibited the pressor response of acute hypoxia. This dose-response curve was shifted leftward when the effects of HMR1766 were investigated in isolated lungs from chronic hypoxic animals for 21 days at 10% oxygen. Mice exposed for 21 or 35 days to chronic hypoxia developed PH, right heart hypertrophy, and pulmonary vascular remodeling. Treatment with HMR1766 (10 mg x kg(-1) x day(-1)), after full establishment of PH from day 21 to day 35, significantly reduced PH, as measured continuously by telemetry. In addition, right ventricular (RV) hypertrophy and structural remodeling of the lung vasculature were reduced. Pharmacological activation of oxidized sGC partially reverses hemodynamic and structural changes in chronic hypoxia-induced experimental PH.
- Published
- 2009
- Full Text
- View/download PDF
25. Heme oxygenase-2 and large-conductance Ca2+-activated K+ channels: lung vascular effects of hypoxia.
- Author
-
Roth M, Rupp M, Hofmann S, Mittal M, Fuchs B, Sommer N, Parajuli N, Quanz K, Schubert D, Dony E, Schermuly RT, Ghofrani HA, Sausbier U, Rutschmann K, Wilhelm S, Seeger W, Ruth P, Grimminger F, Sausbier M, and Weissmann N
- Subjects
- Animals, Hypertension, Pulmonary pathology, Hypertension, Pulmonary physiopathology, Hypoxia pathology, In Vitro Techniques, Lung pathology, Mice, Microscopy, Fluorescence, Muscle, Smooth, Vascular pathology, Muscle, Smooth, Vascular physiopathology, Polymerase Chain Reaction, Pulmonary Alveoli pathology, Pulmonary Wedge Pressure physiology, RNA, Messenger genetics, Heme Oxygenase (Decyclizing) genetics, Heme Oxygenase (Decyclizing) physiology, Hypoxia physiopathology, Large-Conductance Calcium-Activated Potassium Channel alpha Subunits genetics, Large-Conductance Calcium-Activated Potassium Channel alpha Subunits physiology, Lung blood supply, Pulmonary Alveoli blood supply, Vasoconstriction physiology
- Abstract
Rationale: Hypoxic pulmonary vasoconstriction (HPV) is an important mechanism by which pulmonary gas exchange is optimized by the adaptation of blood flow to alveolar ventilation. In chronic hypoxia, in addition to HPV a vascular remodeling process leads to pulmonary hypertension. A complex of heme oxygenase-2 (HO-2) and the BK channel has been suggested as a universal oxygen sensor system., Objectives: We investigated whether this complex serves as an oxygen sensor for the vascular effects of alveolar hypoxia in the lung., Methods: The investigations were performed in chronically hypoxic mice, in isolated perfused and ventilated lungs, and on the cellular level, including HO-2- and BK-channel deficient mice., Measurements and Main Results: Immunohistochemical analysis of mouse lungs identified HO-2 mainly in pulmonary arteries, the bronchial epithelium, and alveolar epithelial cells. BK channel alpha-subunit (BKalpha) immunoreactivity was found primarily in the bronchial and vascular smooth muscle layer. Immunofluorescence staining and coimmunoprecipitation suggested only a weak complexation of HO-2 and BKalpha in pulmonary arterial smooth muscle cells. The strength of acute and sustained HPV, determined in isolated perfused and ventilated lungs, was not different among wild-type, HO-2-deficient, and BKalpha-deficient mice. Exposure of mice to 3 weeks of chronic hypoxia resulted in a slight down-regulation of HO-2 and no alteration in BKalpha expression. The degree of pulmonary hypertension that developed, quantified on the basis of right ventricular pressure, right-heart hypertrophy, and the degree of muscularization of precapillary pulmonary arteries, was not different among wild-type, HO-2-deficient, and BKalpha-deficient mice., Conclusions: It is demonstrated that neither deletion of HO-2 nor BK channels affect acute, sustained, and chronic vascular responses to alveolar hypoxia in the lung.
- Published
- 2009
- Full Text
- View/download PDF
26. Epoxyeicosatrienoic acids and the soluble epoxide hydrolase are determinants of pulmonary artery pressure and the acute hypoxic pulmonary vasoconstrictor response.
- Author
-
Keserü B, Barbosa-Sicard E, Popp R, Fisslthaler B, Dietrich A, Gudermann T, Hammock BD, Falck JR, Weissmann N, Busse R, and Fleming I
- Subjects
- Animals, Blood Pressure, Cytochrome P-450 Enzyme Inhibitors, Cytochrome P-450 Enzyme System metabolism, In Vitro Techniques, Mice, Pulmonary Circulation drug effects, TRPC Cation Channels physiology, TRPC6 Cation Channel, Vasoconstrictor Agents pharmacology, rho-Associated Kinases metabolism, 8,11,14-Eicosatrienoic Acid analogs & derivatives, 8,11,14-Eicosatrienoic Acid pharmacology, Eicosanoids physiology, Epoxide Hydrolases physiology, Hypoxia physiopathology, Pulmonary Artery physiology, Pulmonary Circulation physiology, Vasoconstriction physiology
- Abstract
Recent findings have indicated a role for cytochrome P-450 (CYP) epoxygenase-derived epoxyeicosatrienoic acids (EETs) in acute hypoxic pulmonary vasoconstriction (HPV). Given that the intracellular concentration of EETs is determined by the soluble epoxide hydrolase (sEH), we assessed the influence of the sEH and 11,12-EET on pulmonary artery pressure and HPV in the isolated mouse lung. In lungs from wild-type mice, HPV was significantly increased by sEH inhibition, an effect abolished by pretreatment with CYP epoxygenase inhibitors and the EET antagonist 14,15-EEZE. HPV and EET production were greater in lungs from sEH(-/-) mice than from wild-type mice and sEH inhibition had no further effect on HPV, while MSPPOH and 14,15-EEZE decreased the response. 11,12-EET increased pulmonary artery pressure in a concentration-dependent manner and enhanced HPV via a Rho-dependent mechanism. Both 11,12-EET and hypoxia elicited the membrane translocation of a transient receptor potential (TRP) C6-V5 fusion protein, the latter effect was sensitive to 14,15-EEZE. Moreover, while acute hypoxia and 11,12-EET increased pulmonary pressure in lungs from TRPC6(+/-) mice, lungs from TRPC6(-/-) mice did not respond to either stimuli. These data demonstrate that CYP-derived EETs are involved in HPV and that EET-induced pulmonary contraction under normoxic and hypoxic conditions involves a TRPC6-dependent pathway.
- Published
- 2008
- Full Text
- View/download PDF
27. Regulation of hypoxic pulmonary vasoconstriction: basic mechanisms.
- Author
-
Sommer N, Dietrich A, Schermuly RT, Ghofrani HA, Gudermann T, Schulz R, Seeger W, Grimminger F, and Weissmann N
- Subjects
- 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, Hypoxia, Vasoconstriction
- 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.
- Published
- 2008
- Full Text
- View/download PDF
28. Nitric-oxide-mediated zinc release: a new (modulatory) pathway in hypoxic pulmonary vasoconstriction.
- Author
-
Weissmann N
- Subjects
- Acute Disease, Animals, Calcium Signaling, Cyclic GMP physiology, Endothelial Cells physiology, Humans, Metallothionein deficiency, Metallothionein physiology, Mice, Mice, Knockout, Models, Biological, Nitrosation, Protein Kinase C physiology, Protein Processing, Post-Translational, Pulmonary Artery physiopathology, Rats, Vasoconstriction physiology, Hypoxia physiopathology, Nitric Oxide physiology, Signal Transduction physiology, Vascular Resistance physiology, Zinc physiology
- Published
- 2008
- Full Text
- View/download PDF
29. Hypoxia-driven mechanisms in lung biology and disease: a new review series of the ERS Lung Science Conference.
- Author
-
Weissmann N
- Subjects
- Altitude Sickness physiopathology, Humans, Hypoxia-Inducible Factor 1 physiology, Oxygen metabolism, Hypoxia physiopathology, Lung Diseases physiopathology, Pulmonary Alveoli physiology
- Published
- 2008
- Full Text
- View/download PDF
30. Direct eicosanoid profiling of the hypoxic lung by comprehensive analysis via capillary liquid chromatography with dual online photodiode-array and tandem mass-spectrometric detection.
- Author
-
Kiss L, Röder Y, Bier J, Weissmann N, Seeger W, and Grimminger F
- Subjects
- Animals, Calibration, Eicosanoids chemistry, Molecular Structure, Rabbits, Solvents, Tandem Mass Spectrometry instrumentation, Chromatography, Liquid instrumentation, Chromatography, Liquid methods, Eicosanoids analysis, Hypoxia metabolism, Lung metabolism, Online Systems instrumentation, Tandem Mass Spectrometry methods
- Abstract
Eicosanoids are arachidonic acid-derived mediators, with partly contradictory, incompletely elucidated actions. Thus, epoxyeicosatrienoic acids (EETs) are controversially discussed as putative vasodilatative endothelium-derived hyperpolarizing factors in the cardiovascular compartment but reported as vasoconstrictors in the lung. Inconsistent findings concerning eicosanoid physiology may be because previous methods were lacking sensitivity, identification reliability, and/or have focused on special eicosanoid groups only, ignoring the overall mediator context, and thus limiting the correlation accuracy between autacoid formation and bioactivity profile. Therefore, we developed an approach which enables the simultaneous assessment of 44 eicosanoids, including all representatives of the arachidonic acid cascade, i.e., cytochrome P450, lipoxygenase, cyclooxygenase products, and free isoprostanes as in vivo markers of oxidative stress, in one 50-minute chromatographic run. The approach combines (i) source-specific sample extraction, (ii) rugged isocratic and high-sensitivity capillary liquid-chromatographic separation, and (iii) reliable dual online photodiode-array and electrospray ionization tandem mass-spectrometric identification and quantitation. High sensitivity with limits of quantification in the femtogram range was achieved by use of capillary columns with typical high peak efficiency, due to small inner diameters, and virtually complete substance transfer to the mass spectrometer, due to flow rates in the low microliter range, instead of large inner diameter columns with low chromatographic signal and only partial analyte transfer employed by previous methods. This expeditious, global and sensitive technique provides the prerequisite for new, accurate insights regarding the physiology of specific mediators, for example EETs, in the context of all relevant vasoactive autacoids under varying conditions of oxidative stress by direct comparison of all eicosanoid generation profiles. Indeed, application of comprehensive "eicoprofiling" to hypoxically ventilated rabbit lungs revealed at a glance the enhanced biosynthesis of free EETs in the overall mediator generation context, thus suggesting their hypothetical contribution to hypoxic pulmonary vasoconstriction.
- Published
- 2008
- Full Text
- View/download PDF
31. Sildenafil in hypoxic pulmonary hypertension potentiates a compensatory up-regulation of NO-cGMP signaling.
- Author
-
Kirsch M, Kemp-Harper B, Weissmann N, Grimminger F, and Schmidt HH
- Subjects
- Animals, Hypertension, Pulmonary physiopathology, Hypoxia physiopathology, Lung drug effects, Lung metabolism, Lung physiopathology, Male, Mice, Mice, Inbred C57BL, Phosphorylation, Purines pharmacology, Sildenafil Citrate, Cyclic GMP metabolism, Hypertension, Pulmonary metabolism, Hypoxia metabolism, Nitric Oxide metabolism, Phosphodiesterase Inhibitors pharmacology, Piperazines pharmacology, Signal Transduction drug effects, Sulfones pharmacology, Up-Regulation
- Abstract
The availability of inhibitors of cGMP-specific phosphodiesterase 5 (PDE 5), such as sildenafil, has revolutionized the treatment of pulmonary hypertension (PH). Sildenafil may exert its protective effects in a mechanism-based fashion by targeting a pathophysiologically attenuated NO-cGMP signaling pathway. To elucidate this, we analyzed changes in the pulmonary expression and activity of key enzymes of NO-cGMP signaling as well as the functional pulmonary responses to sildenafil in the 5 or 21 day hypoxia mouse model of PH. Surprisingly, we found doubled NO synthase (NOS) II and III levels, no evidence for attenuated NO bioavailability as evidenced by the nitrosative/oxidative stress marker protein nitro tyrosine, and no changes in the expression and activity of the NO receptor, soluble guanylyl cyclase (sGC). PDE 5 was either unchanged at day 5 or, after 21 days of hypoxia, even significantly decreased along with unchanged activity. Biochemically, these changes were mirrored by increased cGMP spillover into the lung perfusate and cGMP-dependent phosphorylation of the vasodilator-stimulated phosphoprotein, VASP. Sildenafil further augmented cGMP and phospho-VASP levels in lungs of mice exposed for 5 or 21 days and decreased pulmonary arterial pressure in mice after 5 days but not 21 days of hypoxia. In conclusion, NO-cGMP signaling is compensatorily up-regulated in the hypoxic mouse model of PH, and sildenafil further augments this pathway to functionally alleviate pulmonary vasoconstriction.
- Published
- 2008
- Full Text
- View/download PDF
32. Hypoxia-induced pulmonary hypertension: different impact of iloprost, sildenafil, and nitric oxide.
- Author
-
Weissmann N, Gerigk B, Kocer O, Nollen M, Hackemack S, Ghofrani HA, Schermuly RT, Butrous G, Schulz A, Roth M, Seeger W, and Grimminger F
- Subjects
- Analysis of Variance, Animals, Dose-Response Relationship, Drug, Drug Therapy, Combination, Hypertension, Pulmonary etiology, Hypertension, Pulmonary physiopathology, Iloprost administration & dosage, Iloprost pharmacology, Nitric Oxide administration & dosage, Nitric Oxide pharmacology, Piperazines administration & dosage, Piperazines pharmacology, Purines administration & dosage, Purines pharmacology, Rabbits, Sildenafil Citrate, Sulfones administration & dosage, Sulfones pharmacology, Treatment Outcome, Vasodilator Agents administration & dosage, Hypertension, Pulmonary drug therapy, Hypoxia complications, Vascular Resistance drug effects, Vasodilator Agents pharmacology
- Abstract
Objectives: Chronic alveolar hypoxia induces pulmonary hypertension, evident from elevated pulmonary artery pressure (PAP), pulmonary vascular resistance, right ventricular hypertrophy (RVH), and increased muscularization of the pulmonary vasculature. Additionally, the vasoconstrictor response to acute hypoxia (HPV) may be reduced in the remodeled vasculature. However, no direct comparison of different treatments on the various parameters characterizing pulmonary hypertension has been performed yet. Against this background, we compared the effects of inhaled NO, infused iloprost, a stable prostacyclin analogue, and oral sildenafil, a phosphodiesterase 5 inhibitor, on hypoxia-induced pulmonary hypertension., Methods: Exposure of rabbits to chronic hypoxia (FiO(2)=0.10) for 42 days. Treatment with infused iloprost, oral sildenafil, and inhaled nitric oxide., Results: We quantified PAP, pulmonary vascular resistance, RVH, vascular remodeling, vasoreactivity, and the strength of HPV. Chronic hypoxia resulted in an increase in (a) the right ventricle/(left ventricle+septum) ratio from 0.26+/-0.01 to 0.44+/-0.01, (b) PAP, and (c) the degree of muscularization from 14.0+/-4.0% to 43.5+/-5.3%. Treatment with iloprost and sildenafil, but not with NO, prevented the increase in muscularization. In contrast, RVH was strongly inhibited by sildenafil, whereas NO had some minor, and iloprost had no effect. Only iloprost reduced PAP compared to NO and sildenafil. The downregulation of HPV was abrogated only by NO., Conclusion: We demonstrated (a) that the parameters characterizing hypoxia-induced pulmonary hypertension are not functionally linked, (b) that the downregulation of HPV under chronic hypoxia can be prevented by inhaled NO but not by sildenafil and iloprost, and (c) that iloprost is particularly effective in preventing vascular remodeling and sildenafil in preventing RVH.
- Published
- 2007
- Full Text
- View/download PDF
33. Hypoxia-dependent regulation of nonphagocytic NADPH oxidase subunit NOX4 in the pulmonary vasculature.
- Author
-
Mittal M, Roth M, König P, Hofmann S, Dony E, Goyal P, Selbitz AC, Schermuly RT, Ghofrani HA, Kwapiszewska G, Kummer W, Klepetko W, Hoda MA, Fink L, Hänze J, Seeger W, Grimminger F, Schmidt HH, and Weissmann N
- Subjects
- Animals, Cell Division, Cells, Cultured drug effects, Cells, Cultured enzymology, Chronic Disease, Drug Design, Endoplasmic Reticulum enzymology, Enzyme Induction, Female, Humans, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary etiology, Hypertension, Pulmonary physiopathology, Hypertrophy, Hypoxia complications, Hypoxia physiopathology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Lung blood supply, Male, Membrane Glycoproteins analysis, Myocytes, Smooth Muscle enzymology, Myocytes, Smooth Muscle pathology, NADPH Oxidase 2, NADPH Oxidase 4, NADPH Oxidases analysis, NADPH Oxidases biosynthesis, NADPH Oxidases genetics, Nitric Oxide physiology, Organ Specificity, Oxygen metabolism, Oxygen pharmacology, Protein Subunits, Pulmonary Artery cytology, Pulmonary Artery enzymology, RNA Interference, RNA, Messenger biosynthesis, RNA, Small Interfering pharmacology, Superoxides metabolism, Transforming Growth Factor beta1 physiology, Tunica Media enzymology, Tunica Media pathology, Hypertension, Pulmonary enzymology, Hypoxia enzymology, NADPH Oxidases physiology
- Abstract
Nonphagocytic NADPH oxidases have recently been suggested to play a major role in the regulation of physiological and pathophysiological processes, in particular, hypertrophy, remodeling, and angiogenesis in the systemic circulation. Moreover, NADPH oxidases have been suggested to serve as oxygen sensors in the lung. Chronic hypoxia induces vascular remodeling with medial hypertrophy leading to the development of pulmonary hypertension. We screened lung tissue for the expression of NADPH oxidase subunits. NOX1, NOXA1, NOXO1, p22phox, p47phox, p40phox, p67phox, NOX2, and NOX4 were present in mouse lung tissue. Comparing mice maintained for 21 days under hypoxic (10% O(2)) or normoxic (21% O(2)) conditions, an upregulation exclusively of NOX4 mRNA was observed under hypoxia in homogenized lung tissue, concomitant with increased levels in microdissected pulmonary arterial vessels. In situ hybridization and immunohistological staining for NOX4 in mouse lungs revealed a localization of NOX4 mRNA and protein predominantly in the media of small pulmonary arteries, with increased labeling intensities after chronic exposure to hypoxia. In isolated pulmonary arterial smooth muscle cells (PASMCs), NOX4 was localized primarily to the perinuclear space and its expression levels were increased after exposure to hypoxia. Treatment of PASMCs with siRNA directed against NOX4 decreased NOX4 mRNA levels and reduced PASMC proliferation as well as generation of reactive oxygen species. In lungs from patients with idiopathic pulmonary arterial hypertension (IPAH), expression levels of NOX4, which was localized in the vessel media, were 2.5-fold upregulated. These results support an important role for NOX4 in the vascular remodeling associated with development of pulmonary hypertension.
- Published
- 2007
- Full Text
- View/download PDF
34. Classical transient receptor potential channel 6 (TRPC6) is essential for hypoxic pulmonary vasoconstriction and alveolar gas exchange.
- Author
-
Weissmann N, Dietrich A, Fuchs B, Kalwa H, Ay M, Dumitrascu R, Olschewski A, Storch U, Mederos y Schnitzler M, Ghofrani HA, Schermuly RT, Pinkenburg O, Seeger W, Grimminger F, and Gudermann T
- Subjects
- Acute Disease, Animals, Cations chemistry, Cations metabolism, Diglycerides metabolism, Gene Expression, Hypoxia genetics, Lung Diseases genetics, Membrane Potentials, Mice, Mice, Knockout, TRPC Cation Channels deficiency, TRPC Cation Channels genetics, TRPC6 Cation Channel, Aorta metabolism, Hypoxia metabolism, Lung Diseases metabolism, Pulmonary Gas Exchange, TRPC Cation Channels metabolism, Vasoconstriction
- Abstract
Regional alveolar hypoxia causes local vasoconstriction in the lung, shifting blood flow from hypoxic to normoxic areas, thereby maintaining gas exchange. This mechanism is known as hypoxic pulmonary vasoconstriction (HPV). Disturbances in HPV can cause life-threatening hypoxemia whereas chronic hypoxia triggers lung vascular remodeling and pulmonary hypertension. The signaling cascade of this vitally important mechanism is still unresolved. Using transient receptor potential channel 6 (TRPC6)-deficient mice, we show that this channel is a key regulator of acute HPV as this regulatory mechanism was absent in TRPC6(-/-) mice whereas the pulmonary vasoconstrictor response to the thromboxane mimetic U46619 was unchanged. Accordingly, induction of regional hypoventilation resulted in severe arterial hypoxemia in TRPC6(-/-) but not in WT mice. This effect was mirrored by a lack of hypoxia-induced cation influx and currents in smooth-muscle cells from precapillary pulmonary arteries (PASMC) of TRPC6(-/-) mice. In both WT and TRPC6(-/-) PASMC hypoxia caused diacylglycerol (DAG) accumulation. DAG seems to exert its action via TRPC6, as DAG kinase inhibition provoked a cation influx only in WT but not in TRPC6(-/-) PASMC. Notably, chronic hypoxia-induced pulmonary hypertension was independent of TRPC6 activity. We conclude that TRPC6 plays a unique and indispensable role in acute hypoxic pulmonary vasoconstriction. Manipulation of TRPC6 function may thus offer a therapeutic strategy for the control of pulmonary hemodynamics and gas exchange.
- Published
- 2006
- Full Text
- View/download PDF
35. Hypoxia- and non-hypoxia-related pulmonary hypertension - established and new therapies.
- Author
-
Ghofrani HA, Voswinckel R, Reichenberger F, Weissmann N, Schermuly RT, Seeger W, and Grimminger F
- Subjects
- Animals, Antihypertensive Agents therapeutic use, Humans, Hypertension, Pulmonary drug therapy, Hypoxia drug therapy, Lung Diseases, Interstitial complications, Lung Diseases, Interstitial drug therapy, Pulmonary Disease, Chronic Obstructive complications, Pulmonary Disease, Chronic Obstructive drug therapy, Thrombosis complications, Thrombosis drug therapy, Vasodilator Agents therapeutic use, Hypertension, Pulmonary etiology, Hypoxia complications
- Abstract
Pulmonary hypertension can occur as an isolated disease affecting the lung vessels only, in association with underlying hypoxic lung disorders, or due to chronic thromboembolic disease. Pulmonary hypertension caused by pulmonary venous congestion will not be focused on in this review. Regardless of the underlying disease, chronic cor pulmonale is associated with progressive clinical deterioration and a poor prognosis in most cases. The aim of specific therapies for pulmonary hypertension is to reduce pulmonary vascular resistance and thereby improve right ventricular function. Currently, three classes of drugs (prostanoids, endothelin receptor antagonists, and phosphodiesterase-5 inhibitors) are approved for the treatment of pulmonary arterial hypertension (PAH) in a defined patient population (group I according to the recent WHO classification). However, these medications may also lower pulmonary vascular resistance in patients with associated lung diseases (e.g. chronic obstructive pulmonary disease or lung fibrosis) and significant pulmonary hypertension, for whom these drugs are not yet approved. As non-selective vasodilators may induce gas-exchange disturbances, which preclude their long term use in these patients, such substances should be avoided in the hypoxemic patient. In this article we provide an update of the current understanding of hypoxia- and non-hypoxia-related pulmonary hypertension, addressing both the pathophysiological understanding of different disease aetiologies as well as the therapeutic options currently available.
- Published
- 2006
- Full Text
- View/download PDF
36. Increased protein arginine methylation in chronic hypoxia: role of protein arginine methyltransferases.
- Author
-
Yildirim AO, Bulau P, Zakrzewicz D, Kitowska KE, Weissmann N, Grimminger F, Morty RE, and Eickelberg O
- Subjects
- Animals, Isoenzymes metabolism, Male, Methylation, Mice, Mice, Inbred BALB C, Protein-Arginine N-Methyltransferases metabolism, Arginine analogs & derivatives, Arginine metabolism, Hypoxia enzymology, Lung enzymology, Protein-Arginine N-Methyltransferases physiology
- Abstract
Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of nitric oxide synthesis. ADMA is generated by catabolism of proteins containing methylated arginine residues, and its levels are correlated with endothelial dysfunction in systemic cardiovascular diseases. Arginine methylation of cellular proteins is catalyzed by protein arginine methyltransferases (PRMT). The expression and localization of PRMT in the lung has not been addressed. Here, we sought to analyze the expression of PRMT isoforms in the lung and to determine whether PRMT expression is altered during exposure to chronic hypoxia (10% oxygen). Adult mice were exposed to hypoxia for up to 3 wk, and lung tissues were harvested and processed for RT-PCR, Western blotting, immunohistochemistry, and determination of tissue ADMA levels. All PRMT isoforms investigated were detected at the mRNA and protein level in mouse lung, and were localized primarily to the bronchial and alveolar epithelium. In lungs of mice subjected to chronic hypoxia, PRMT2 mRNA and protein levels were up-regulated, whereas the expression of all other PRMT isoforms remained unchanged. This was mainly due to increased expression of PRMT2 in alveolar type II cells, which did not express detectable levels of PRMT2 under normoxic conditions. Consistent with these observations, lung ADMA levels and ADMA/l-Arginine ratios were increased under hypoxic conditions. These results demonstrate that PRMTs are expressed and functional in the lung, and that hypoxia is a potent regulator of PRMT2 expression and lung ADMA concentrations. These data suggest that structural and functional changes caused by hypoxia may be linked to ADMA metabolism.
- Published
- 2006
- Full Text
- View/download PDF
37. Hypoxia in lung vascular biology and disease.
- Author
-
Weissmann N, Grimminger F, and Seeger W
- Subjects
- Adaptation, Physiological, Animals, Humans, Hypertension, Pulmonary physiopathology, Lung physiopathology, Pulmonary Gas Exchange, Vasoconstriction physiology, Blood-Air Barrier, Hypoxia metabolism, Lung physiology, Pulmonary Circulation
- Published
- 2006
- Full Text
- View/download PDF
38. Oxygen sensors in hypoxic pulmonary vasoconstriction.
- Author
-
Weissmann N, Sommer N, Schermuly RT, Ghofrani HA, Seeger W, and Grimminger F
- Subjects
- Adenosine Triphosphate metabolism, Animals, Calcium metabolism, Cytochrome P-450 Enzyme System metabolism, Heme Oxygenase (Decyclizing) metabolism, Humans, Hypertension, Pulmonary metabolism, Mitochondria metabolism, Reactive Oxygen Species metabolism, Vasoconstriction physiology, Hypoxia metabolism, Lung metabolism, Oxygen metabolism
- Abstract
Hypoxic pulmonary vasoconstriction (HPV) is an essential mechanism adapting lung perfusion to regional ventilation. Perturbations to HPV, such as those occurring in pneumonia, acute respiratory distress syndrome and liver failure, can result in arterial hypoxemia. Under conditions of general hypoxia, HPV increases pulmonary vascular resistance and thus causes acute pulmonary hypertension. Despite intensive research, the underlying mechanisms of HPV have not been fully elucidated. Deciphering signalling pathways that result in HPV could suggest novel approaches to address a failure of HPV, as well as for the treatment of pulmonary hypertension associated with HPV. Within this context, this review focuses on current concepts in the oxygen sensing mechanisms that underlie HPV.
- Published
- 2006
- Full Text
- View/download PDF
39. Impact of mitochondria and NADPH oxidases on acute and sustained hypoxic pulmonary vasoconstriction.
- Author
-
Weissmann N, Zeller S, Schäfer RU, Turowski C, Ay M, Quanz K, Ghofrani HA, Schermuly RT, Fink L, Seeger W, and Grimminger F
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Animals, Electron Transport Complex I physiology, Electron Transport Complex II physiology, Electron Transport Complex III physiology, Electron Transport Complex IV physiology, Female, Hypoxia physiopathology, In Vitro Techniques, Male, Membrane Glycoproteins genetics, Mice, Mice, Inbred C57BL, NADPH Oxidase 2, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases genetics, Protein Isoforms antagonists & inhibitors, Protein Isoforms physiology, Pulmonary Gas Exchange, Rabbits, Superoxides metabolism, Vasoconstrictor Agents pharmacology, Hypoxia metabolism, Lung blood supply, Mitochondria physiology, NADPH Oxidases physiology, Oxygen physiology, Vasoconstriction drug effects
- Abstract
Hypoxic pulmonary vasoconstriction (HPV) matches lung perfusion with ventilation to optimize pulmonary gas exchange. However, it remains unclear whether acute HPV (occurring within seconds) and the vasoconstrictor response to sustained alveolar hypoxia (developing over several hours) are triggered by identical mechanisms. We investigated the effect of mitochondrial and NADPH oxidase inhibitors on both phases of HPV in intact rabbit lungs. These studies revealed that the sustained HPV is largely dependent on mitochondrial complex I and totally dependent on complex IV, whereas NADPH oxidase dependence was only observed for acute HPV. These findings were reinforced by an alternative approach employing lungs from mice deficient in the NADPH oxidase subunit p 47(phox). In these mice (which lack a subunit suggested to be important for the function of most NADPH oxidase isoforms), but not in gp 91(phox)-deficient mice (which represent only one isoform of NADPH oxidases), acute HPV was significantly reduced, while non-hypoxia-induced vasoconstrictions elicited by the thromboxane mimetic U46619 were not affected. We concluded that the acute phase and the sustained phase of HPV are differentially regulated, with NADPH oxidase activity predominating in the acute phase, while a strong dependence on mitochondrial participation was observed for the second phase.
- Published
- 2006
- Full Text
- View/download PDF
40. Hypoxic pulmonary vasoconstriction--triggered by an increase in reactive oxygen species?
- Author
-
Weissmann N, Schermuly RT, Ghofrani HA, Hänze J, Goyal P, Grimminger F, and Seeger W
- Subjects
- Animals, Pulmonary Artery physiopathology, Rabbits, Reactive Oxygen Species antagonists & inhibitors, Hypoxia metabolism, Hypoxia physiopathology, Pulmonary Artery metabolism, Reactive Oxygen Species metabolism, Vasoconstriction physiology
- Abstract
Hypoxic pulmonary vasoconstriction (HPV) is an essential mechanism of the lung that matches perfusion to ventilation in order to optimize pulmonary gas exchange. Despite intensive research, the underlying mechanism has not yet been fully elucidated. Reactive oxygen species (ROS) have been proposed as key mediators of HPV. However, there is ongoing discussion as to whether ROS really contribute to HPV regulation and if so, whether an increase or a decrease in ROS occurs during alveolar hypoxia. In this overview, we summarize our data that have led us to conclude that alveolar hypoxia induces an increase in superoxide and subsequently H2O2, and thus elicits HPV. This conclusion is drawn from investigations employing various inhibitors that interfere with ROS in isolated buffer-perfused rabbit lungs challenged with 10-minute periods of alveolar hypoxia. Targeting possible sources of a hypoxia-induced increase in ROS, our data are only partially in accordance with the hypothesis that mitochondria are the hypoxia-dependent ROS generators, and suggest NADPH oxidases as an alternative source. From measurements of intracellular and exhaled H2O2, we hypothesize that total lung ROS release is reduced in alveolar hypoxia, but that in specialized cells or sub-cellular structures an increased ROS release may occur, triggering HPV.
- Published
- 2006
41. Impact of HIF-1alpha and HIF-2alpha on proliferation and migration of human pulmonary artery fibroblasts in hypoxia.
- Author
-
Eul B, Rose F, Krick S, Savai R, Goyal P, Klepetko W, Grimminger F, Weissmann N, Seeger W, and Hänze J
- Subjects
- Actins metabolism, Apoptosis, Basic Helix-Loop-Helix Transcription Factors, Cell Cycle, Cell Proliferation, Cells, Cultured, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Oxygen metabolism, RNA Interference, Transcription Factors genetics, Cell Movement, Fibroblasts cytology, Fibroblasts metabolism, Hypoxia metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Pulmonary Artery cytology, Transcription Factors metabolism
- Abstract
Proliferation of adventitial fibroblasts of small intrapulmonary arteries (FBPA) has been disclosed as an early event in the development of pulmonary hypertension and cor pulmonale in response to hypoxia. We investigated the role of hypoxia-inducible transcription factors (HIF) in human FBPA exposed to hypoxia. Primary cultures of FBPA displayed a strong mitogenic response to 24 h hypoxia, whereas the rate of apoptosis was significantly suppressed. In addition, the migration of FBPA was strongly increased under hypoxic conditions but not the expression of alpha-smooth muscle actin. Hypoxia induced a marked up-regulation (protein level) of both HIF-1alpha and HIF-2alpha, alongside with nuclear translocation of these transcription factors. Specific inhibition of either HIF-1alpha or HIF-2alpha was achieved by RNA interference technology, as proven by HIF-1alpha and HIF-2alpha mRNA and protein analysis and expression analysis of HIF downstream target genes. With the use of this approach, the hypoxia-induced proliferative response of the FBPA was found to be solely HIF-2alpha dependent, whereas the migratory response was significantly reduced by both HIF-1alpha and HIF-2alpha interference. In conclusion, HIF up-regulation is essential for hypoxic cellular responses in human pulmonary artery adventitial fibroblasts such as proliferation and migration, mimicking the pulmonary hypertensive phenotype in vivo. Differential HIF subtype dependency was noted, with HIF-2alpha playing a predominant role, which may offer future intervention strategies.
- Published
- 2006
- Full Text
- View/download PDF
42. Expression profiling of laser-microdissected intrapulmonary arteries in hypoxia-induced pulmonary hypertension.
- Author
-
Kwapiszewska G, Wilhelm J, Wolff S, Laumanns I, Koenig IR, Ziegler A, Seeger W, Bohle RM, Weissmann N, and Fink L
- Subjects
- Animals, Gene Expression Profiling, Hypertension, Pulmonary etiology, Hypoxia complications, Laser Therapy, Male, Mice, Mice, Inbred BALB C, Microdissection, Cytokines metabolism, Hypertension, Pulmonary metabolism, Hypertension, Pulmonary pathology, Hypoxia metabolism, Hypoxia pathology, Pulmonary Artery metabolism, Pulmonary Artery pathology
- Abstract
Background: Chronic hypoxia influences gene expression in the lung resulting in pulmonary hypertension and vascular remodelling. For specific investigation of the vascular compartment, laser-microdissection of intrapulmonary arteries was combined with array profiling., Methods and Results: Analysis was performed on mice subjected to 1, 7 and 21 days of hypoxia (FiO2 = 0.1) using nylon filters (1176 spots). Changes in the expression of 29, 38, and 42 genes were observed at day 1, 7, and 21, respectively. Genes were grouped into 5 different classes based on their time course of response. Gene regulation obtained by array analysis was confirmed by real-time PCR. Additionally, the expression of the growth mediators PDGF-B, TGF-beta, TSP-1, SRF, FGF-2, TIE-2 receptor, and VEGF-R1 were determined by real-time PCR. At day 1, transcription modulators and ion-related proteins were predominantly regulated. However, at day 7 and 21 differential expression of matrix producing and degrading genes was observed, indicating ongoing structural alterations. Among the 21 genes upregulated at day 1, 15 genes were identified carrying potential hypoxia response elements (HREs) for hypoxia-induced transcription factors. Three differentially expressed genes (S100A4, CD36 and FKBP1a) were examined by immunohistochemistry confirming the regulation on protein level. While FKBP1a was restricted to the vessel adventitia, S100A4 and CD36 were localised in the vascular tunica media., Conclusion: Laser-microdissection and array profiling has revealed several new genes involved in lung vascular remodelling in response to hypoxia. Immunohistochemistry confirmed regulation of three proteins and specified their localisation in vascular smooth muscle cells and fibroblasts indicating involvement of different cells types in the remodelling process. The approach allows deeper insight into hypoxic regulatory pathways specifically in the vascular compartment of this complex organ.
- Published
- 2005
- Full Text
- View/download PDF
43. Congenital erythropoietin over-expression causes "anti-pulmonary hypertensive" structural and functional changes in mice, both in normoxia and hypoxia.
- Author
-
Weissmann N, Manz D, Buchspies D, Keller S, Mehling T, Voswinckel R, Quanz K, Ghofrani HA, Schermuly RT, Fink L, Seeger W, Gassmann M, and Grimminger F
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Animals, Aspirin pharmacology, Blood Pressure, Blood Vessels pathology, Blood Viscosity, Cyclooxygenase Inhibitors pharmacology, Erythropoietin blood, Erythropoietin genetics, Hematocrit, Hypertension, Pulmonary blood, Hypertension, Pulmonary etiology, Hypertension, Pulmonary physiopathology, Hypertrophy, Left Ventricular physiopathology, Hypertrophy, Right Ventricular physiopathology, Lung pathology, Lung physiopathology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Muscle, Smooth, Vascular pathology, Nitric Oxide Synthase antagonists & inhibitors, Up-Regulation, Vascular Resistance drug effects, Vasoconstrictor Agents pharmacology, omega-N-Methylarginine pharmacology, Erythropoietin physiology, Hypertension, Pulmonary prevention & control, Hypoxia physiopathology, Lung blood supply, Vasoconstriction drug effects
- Abstract
Acute alveolar hypoxia causes pulmonary vasoconstriction that matches lung perfusion to ventilation to optimize gas exchange. Chronic alveolar hypoxia induces pulmonary hypertension, characterized by increased muscularization of the pulmonary vasculature and right ventricular hypertrophy. Elevated erythropoietin (EPO) plasma levels increase hematocrit and blood viscosity and may affect structure and function of the pulmonary circulation. To differentiate between the direct effects of hypoxia and those linked to a hypoxia-induced increase in EPO/hematocrit levels, we investigated the lung vasculature in transgenic mice constitutively over-expressing EPO (termed tg6) upon exposure to normoxia and chronic hypoxia. Despite increased hematocrit levels (approximately 0.86),tg6 mice kept in normoxia did not develop selective right ventricular hypertrophy. The portion of vessels with a diameter of 51-95 microm and >155 microm was increased whereas the portion of small vessels (30-50 microm) was decreased. Pulmonary vascular resistance and the strength of hypoxic vasoconstriction measured in isolated perfused lungs were decreased. Vasoconstrictions induced by the thromboxane mimetic U46619 tended to be reduced. After chronic hypoxia (FiO2 = 0.10, 21 days), vascular resistance and vasoconstrictor responses to acute hypoxia and U46619 were reduced in tg6 mice compared to wildtype controls. Chronic hypoxia increased the degree of pulmonary vascular muscularization in wildtype but not in tg6 mice that already exhibited less muscularization in normoxia. In conclusion, congenital over-expression of EPO exerts an "anti-pulmonary hypertensive" effect, both structurally and functionally, particularly obvious upon chronic hypoxia.
- Published
- 2005
- Full Text
- View/download PDF
44. Basic features of hypoxic pulmonary vasoconstriction in mice.
- Author
-
Weissmann N, Akkayagil E, Quanz K, Schermuly RT, Ghofrani HA, Fink L, Hänze J, Rose F, Seeger W, and Grimminger F
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Amylopectin metabolism, Animals, Aspirin pharmacology, Blood Pressure drug effects, Drug Interactions, Enzyme Inhibitors pharmacology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Pulmonary Artery drug effects, Pulmonary Circulation drug effects, Pulmonary Gas Exchange physiology, Species Specificity, Tidal Volume physiology, Vasoconstriction drug effects, Vasoconstrictor Agents pharmacology, omega-N-Methylarginine pharmacology, Amylopectin analogs & derivatives, Blood Pressure physiology, Hypoxia physiopathology, Oxygen metabolism, Pulmonary Artery physiopathology, Vasoconstriction physiology
- Abstract
Hypoxic pulmonary vasoconstriction (HPV) matches lung perfusion with ventilation which tends to optimize pulmonary gas exchange. Investigations using genetically engineered mice represent a promising approach to understand the underlying mechanisms. Our goal was to characterize basic features of HPV in the isolated buffer-perfused and ventilated mouse lung system. HPV was reproducible for several hours when ventilating the lungs with 1% O2 (10 min) alternated with normoxic ventilation periods (21% O2, 15 min). HPV was well elicitable and most constant using Krebs-Henseleit buffer with the addition of hydroxyethylamylopectin as an oncotic agent. Inhibition of both lung NO and prostanoid formation amplified HPV in an over-additive fashion. HPV was higher in BALB/c mive as compared to C57BL/6 mice, and was approximately threefold enhanced under positive pressure ventilation as compared to negative pressure ventilation. A three hour hypoxic ventilation period resulted in a biphasic vasoconstrictor response with loss of posthypoxic vasodilatation. In summary, we have characterised HPV and established an experimental set-up optimized for investigation of the basic mechanisms of HPV in mice.
- Published
- 2004
- Full Text
- View/download PDF
45. Effects of mitochondrial inhibitors and uncouplers on hypoxic vasoconstriction in rabbit lungs.
- Author
-
Weissmann N, Ebert N, Ahrens M, Ghofrani HA, Schermuly RT, Hänze J, Fink L, Rose F, Conzen J, Seeger W, and Grimminger F
- Subjects
- 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid pharmacology, Animals, Antifungal Agents pharmacology, Antimycin A pharmacology, Dose-Response Relationship, Drug, In Vitro Techniques, Lung cytology, Methacrylates, Nitric Oxide metabolism, Rabbits, Rotenone pharmacology, Thiazoles pharmacology, omega-N-Methylarginine pharmacology, Enzyme Inhibitors pharmacology, Hypoxia metabolism, Lung physiology, Mitochondria drug effects, Mitochondria metabolism, Uncoupling Agents pharmacology, Vasoconstriction physiology
- Abstract
Hypoxic pulmonary vasoconstriction (HPV) matches lung perfusion to ventilation for optimizing pulmonary gas exchange; however, the underlying mechanism has not yet been fully elucidated. Lung nitric oxide (NO) generation appears to be involved in this process. Recently, mitochondria have been proposed as oxygen sensors, with HPV signaling via a hypoxia-induced increase in the generation of reactive oxygen species derived from mitochondrial complex III and escaping through an anion channel into the cytoplasm. In addition, complex II has been suggested to be specifically involved in hypoxia-dependent generation of reactive oxygen species in the lung. We investigated the effects of several mitochondrial inhibitors and uncouplers on the strength of HPV, and asked for their capacity to mimic HPV during normoxia in isolated buffer-perfused rabbit lungs. Specificity of the agents for HPV was tested by comparison of their effects on non-hypoxia-induced vasoconstriction, elicited by the thromboxane mimetic U46619. Interference with NO metabolism was determined by performing parallel studies with blocked lung NO generation and by measurement of exhaled NO. Rotenone, 3-nitroproprionic acid, and myxothiazol dose-dependently inhibited HPV without being mimics of HPV during normoxia. The inhibitory effect of these agents was only partly specific for HPV by comparison with U46619-induced vasoconstriction. During pre-blocked lung NO synthesis, the selectivity for HPV inhibition was increased for rotenone, but largely lost for myxothiazol. 2-tenoyltrifluoroacetone resulted in an unspecific inhibition of HPV as compared with U46619-induced vasoconstriction. 1-methyl-4-phenylpyridinium iodide and 2-heptyl-4-hydroxyquinoline-N-oxide specifically suppressed HPV and increased normoxic vascular tone. Antimycin A suppressed HPV, an effect being specific in lungs with intact NO synthesis and only partly specific while blocking NO. However, this agent did not mimic HPV during normoxia, as may be expected for interference with the mitochondrial electron transport downstream in complex III. The uncouplers 2,4-dinitrophenol (DNP, 10-200 microM) and carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP, 1-3 microM) induced sustained vasoconstriction during normoxia, with enhancement of HPV by DNP at low and suppression of HPV for both agents at high concentrations. The anion channel blocker 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid inhibited HPV and U46619-induced vasoconstriction with identical dose-response curves. These findings suggest that mitochondria are in some manner involved in the regulation of HPV in intact rabbit lungs. The hypothesis that enhanced superoxide leak at complex III of mitochondria represents the underlying mechanism of acute HPV is supported by the rotenone and 2-heptyl-4-hydroxyquinoline-N-oxide data, but partly contradicted by the findings with 1-methyl-4-phenylpyridinium iodide, antimycin A, DNP, and FCCP. Further studies are mandatory to clarify the link between mitochondrial respiratory chain and hypoxic pulmonary vasoconstriction.
- Published
- 2003
- Full Text
- View/download PDF
46. Essential role of complex II of the respiratory chain in hypoxia-induced ROS generation in the pulmonary vasculature.
- Author
-
Paddenberg R, Ishaq B, Goldenberg A, Faulhammer P, Rose F, Weissmann N, Braun-Dullaeus RC, and Kummer W
- Subjects
- Animals, Anti-Bacterial Agents pharmacology, Antihypertensive Agents pharmacology, Antimycin A pharmacology, Chelating Agents pharmacology, Electron Transport physiology, Electron Transport Complex II, Electron Transport Complex III antagonists & inhibitors, Electron Transport Complex IV antagonists & inhibitors, Enzyme Inhibitors pharmacology, Mice, Mice, Inbred Strains, Mitochondria metabolism, Multienzyme Complexes antagonists & inhibitors, Nitric Oxide metabolism, Nitro Compounds, Organ Culture Techniques, Oxidoreductases antagonists & inhibitors, Propionates pharmacology, Sodium Azide pharmacology, Succinate Dehydrogenase antagonists & inhibitors, Thenoyltrifluoroacetone pharmacology, Hypoxia metabolism, Multienzyme Complexes metabolism, Oxidoreductases metabolism, Pulmonary Circulation physiology, Reactive Oxygen Species metabolism, Succinate Dehydrogenase metabolism
- Abstract
In the pulmonary vasculature, the mechanisms responsible for oxygen sensing and the initiation of hypoxia-induced vasoconstriction and vascular remodeling are still unclear. Nitric oxide (NO) and reactive oxygen species (ROS) are discussed as early mediators of the hypoxic response. Here, we describe a quantitative analysis of NO- and ROS-producing cells within the vascular walls of murine lung sections cultured at normoxia or hypoxia. Whereas the number of NO-producing cells was not changed by hypoxia, the number of ROS-generating cells was significantly increased. Addition of specific inhibitors revealed that mitochondria were the source of ROS. The participation of the individual mitochondrial complexes differed in normoxic and hypoxic ROS generation. Whereas normoxic ROS production required complexes I and III, hypoxic ROS generation additionally demanded complex II. Histochemically demonstrable succinate dehydrogenase activity of complex II in the arterial wall decreased during hypoxia. Inhibition of the reversed enzymatic reaction, i.e., fumarate reductase, by application of succinate, specifically abolished hypoxic, but not normoxic, ROS generation. Thus complex II plays an essential role in hypoxic ROS production. Presumably, its catalytic activity switches from succinate dehydrogenase to fumarate reductase at reduced oxygen tension, thereby modulating the directionality of the electron flow.
- Published
- 2003
- Full Text
- View/download PDF
47. Downregulation of hypoxic vasoconstriction by chronic hypoxia in rabbits: effects of nitric oxide.
- Author
-
Weissmann N, Nollen M, Gerigk B, Ardeschir Ghofrani H, Schermuly RT, Gunther A, Quanz K, Fink L, Hänze J, Rose F, Seeger W, and Grimminger F
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Acute Disease, Animals, Breath Tests, Chronic Disease, Disease Models, Animal, Enzyme Inhibitors pharmacology, Female, Hypertension, Pulmonary etiology, Hypertension, Pulmonary pathology, Hypertrophy, Right Ventricular etiology, Hypertrophy, Right Ventricular pathology, Hypoxia complications, Hypoxia drug therapy, In Vitro Techniques, Lung blood supply, Lung pathology, Lung physiopathology, Male, Nitric Oxide administration & dosage, Nitric Oxide metabolism, Nitric Oxide Synthase antagonists & inhibitors, Rabbits, Time, Vasoconstrictor Agents pharmacology, omega-N-Methylarginine pharmacology, Hypertension, Pulmonary physiopathology, Hypoxia physiopathology, Nitric Oxide pharmacology, Vasoconstriction drug effects
- Abstract
Hypoxic pulmonary vasoconstriction (HPV) matches lung perfusion to ventilation for optimizing pulmonary gas exchange. Chronic alveolar hypoxia results in vascular remodeling and pulmonary hypertension. Previous studies have reported conflicting results of the effect of chronic alveolar hypoxia on pulmonary vasoreactivity and the contribution of nitric oxide (NO), which may be related to species and strain differences as well as to the duration of chronic hypoxia. Therefore, we investigated the impact of chronic hypoxia on HPV in rabbits, with a focus on lung NO synthesis. After exposure of the animals to normobaric hypoxia (10% O(2)) for 1 day to 10 wk, vascular reactivity was investigated in ex vivo perfused normoxic ventilated lungs. Chronic hypoxia induced right heart hypertrophy and increased normoxic vascular tone within weeks. The vasoconstrictor response to an acute hypoxic challenge was strongly downregulated within 5 days, whereas the vasoconstrictor response to the thromboxane mimetic U-46619 was maintained. The rapid downregulation of HPV was apparently not linked to changes in the lung vascular NO system, detectable in the exhaled gas and by pharmacological blockage of NO synthesis. Treatment of the animals with long-term inhaled NO reduced right heart hypertrophy and partially maintained the reactivity to acute hypoxia, without any impact on the endogenous NO system being noted. We conclude that chronic hypoxia causes rapid downregulation of acute HPV as a specific event, preceding the development of major pulmonary hypertension and being independent of the lung vascular NO system. Long-term NO inhalation partially maintains the strength of the hypoxic vasoconstrictor response.
- Published
- 2003
- Full Text
- View/download PDF
48. [Vascular effects of alveolar hypoxia--sensing and signal transduction mechanisms].
- Author
-
Weissmann N
- Subjects
- Humans, Hypoxia etiology, Hypoxia physiopathology, Pulmonary Alveoli blood supply, Pulmonary Alveoli physiopathology, Signal Transduction
- Published
- 2002
- Full Text
- View/download PDF
49. Hypoxic pulmonary vasoconstriction: a multifactorial response?
- Author
-
Weissmann N, Grimminger F, Olschewski A, and Seeger W
- Subjects
- Animals, Hypoxia physiopathology, Pulmonary Artery physiopathology, Vasoconstriction
- Published
- 2001
- Full Text
- View/download PDF
50. NO and reactive oxygen species are involved in biphasic hypoxic vasoconstriction of isolated rabbit lungs.
- Author
-
Weissmann N, Winterhalder S, Nollen M, Voswinckel R, Quanz K, Ghofrani HA, Schermuly RT, Seeger W, and Grimminger F
- Subjects
- Animals, Blood Pressure, In Vitro Techniques, Pulmonary Artery physiopathology, Rabbits, Hypoxia physiopathology, Nitric Oxide physiology, Pulmonary Circulation, Reactive Oxygen Species physiology, Vasoconstriction physiology
- Abstract
Hypoxic pulmonary vasoconstriction (HPV) matches lung perfusion with ventilation but may also result in chronic pulmonary hypertension. It has not been clarified whether acute HPV and the response to prolonged alveolar hypoxia are triggered by identical mechanisms. We characterized the vascular response to sustained hypoxic ventilation (3% O(2) for 120-180 min) in isolated rabbit lungs. Hypoxia provoked a biphasic increase in pulmonary arterial pressure (PAP). Persistent PAP elevation was observed after termination of hypoxia. Total blockage of lung nitric oxide (NO) formation by N(G)-monomethyl-L-arginine caused a two- to threefold amplification of acute HPV, the sustained pressor response, and the loss of posthypoxic relaxation. This amplification was only moderate when NO formation was partially blocked by the inducible NO synthase inhibitor S-methylisothiourea. The superoxide scavenger nitro blue tetrazolium and the superoxide dismutase inhibitor triethylenetetramine reduced the initial vasoconstrictor response, the prolonged PAP increase, and the loss of posthypoxic vasorelaxation to a similar extent. The NAD(P)H oxidase inhibitor diphenyleneiodonium nearly fully blocked the late vascular responses to hypoxia in a dose that effected a decrease to half of the acute HPV. In conclusion, as similarly suggested for acute HPV, lung NO synthesis and the superoxide-hydrogen peroxide axis appear to be implicated in the prolonged pressor response and the posthypoxic loss of vasorelaxation in perfused rabbit lungs undergoing 2-3 h of hypoxic ventilation.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.