1
|
Moreno-Domínguez A, Colinas O, Smani T, Ureña J, López-Barneo J. Acute oxygen sensing by vascular smooth muscle cells. Front Physiol 2023; 14:1142354. [PMID: 36935756 PMCID: PMC10020353 DOI: 10.3389/fphys.2023.1142354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
An adequate supply of oxygen (O2) is essential for most life forms on earth, making the delivery of appropriate levels of O2 to tissues a fundamental physiological challenge. When O2 levels in the alveoli and/or blood are low, compensatory adaptive reflexes are produced that increase the uptake of O2 and its distribution to tissues within a few seconds. This paper analyzes the most important acute vasomotor responses to lack of O2 (hypoxia): hypoxic pulmonary vasoconstriction (HPV) and hypoxic vasodilation (HVD). HPV affects distal pulmonary (resistance) arteries, with its homeostatic role being to divert blood to well ventilated alveoli to thereby optimize the ventilation/perfusion ratio. HVD is produced in most systemic arteries, in particular in the skeletal muscle, coronary, and cerebral circulations, to increase blood supply to poorly oxygenated tissues. Although vasomotor responses to hypoxia are modulated by endothelial factors and autonomic innervation, it is well established that arterial smooth muscle cells contain an acute O2 sensing system capable of detecting changes in O2 tension and to signal membrane ion channels, which in turn regulate cytosolic Ca2+ levels and myocyte contraction. Here, we summarize current knowledge on the nature of O2 sensing and signaling systems underlying acute vasomotor responses to hypoxia. We also discuss similarities and differences existing in O2 sensors and effectors in the various arterial territories.
Collapse
Affiliation(s)
- Alejandro Moreno-Domínguez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Olaia Colinas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Tarik Smani
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Juan Ureña
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- *Correspondence: José López-Barneo,
| |
Collapse
|
2
|
Shimoda LA, Suresh K, Undem C, Jiang H, Yun X, Sylvester JT, Swenson ER. Acetazolamide prevents hypoxia-induced reactive oxygen species generation and calcium release in pulmonary arterial smooth muscle. Pulm Circ 2021; 11:20458940211049948. [PMID: 34646499 PMCID: PMC8504243 DOI: 10.1177/20458940211049948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
Upon sensing a reduction in local oxygen partial pressure, pulmonary vessels constrict, a phenomenon known as hypoxic pulmonary vasoconstriction. Excessive hypoxic pulmonary vasoconstriction can occur with ascent to high altitude and is a contributing factor to the development of high-altitude pulmonary edema. The carbonic anhydrase inhibitor, acetazolamide, attenuates hypoxic pulmonary vasoconstriction through stimulation of alveolar ventilation via modulation of acid-base homeostasis and by direct effects on pulmonary vascular smooth muscle. In pulmonary arterial smooth muscle cells (PASMCs), acetazolamide prevents hypoxia-induced increases in intracellular calcium concentration ([Ca2+]i), although the exact mechanism by which this occurs is unknown. In this study, we explored the effect of acetazolamide on various calcium-handling pathways in PASMCs. Using fluorescent microscopy, we tested whether acetazolamide directly inhibited store-operated calcium entry or calcium release from the sarcoplasmic reticulum, two well-documented sources of hypoxia-induced increases in [Ca2+]i in PASMCs. Acetazolamide had no effect on calcium entry stimulated by store-depletion, nor on calcium release from the sarcoplasmic reticulum induced by either phenylephrine to activate inositol triphosphate receptors or caffeine to activate ryanodine receptors. In contrast, acetazolamide completely prevented Ca2+-release from the sarcoplasmic reticulum induced by hypoxia (4% O2). Since these results suggest the acetazolamide interferes with a mechanism upstream of the inositol triphosphate and ryanodine receptors, we also determined whether acetazolamide might prevent hypoxia-induced changes in reactive oxygen species production. Using roGFP, a ratiometric reactive oxygen species-sensitive fluorescent probe, we found that hypoxia caused a significant increase in reactive oxygen species in PASMCs that was prevented by 100 μM acetazolamide. Together, these results suggest that acetazolamide prevents hypoxia-induced changes in [Ca2+]i by attenuating reactive oxygen species production and subsequent activation of Ca2+-release from sarcoplasmic reticulum stores.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Clark Undem
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Xin Yun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - J T Sylvester
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Erik R Swenson
- Division of Pulmonary and Critical Care Medicine, VA Puget Sound Health Care System and University of Washington School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
Generation of Reactive Oxygen Species by Mitochondria. Antioxidants (Basel) 2021; 10:antiox10030415. [PMID: 33803273 PMCID: PMC8001687 DOI: 10.3390/antiox10030415] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/28/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species (ROS) are series of chemical products originated from one or several electron reductions of oxygen. ROS are involved in physiology and disease and can also be both cause and consequence of many biological scenarios. Mitochondria are the main source of ROS in the cell and, particularly, the enzymes in the electron transport chain are the major contributors to this phenomenon. Here, we comprehensively review the modes by which ROS are produced by mitochondria at a molecular level of detail, discuss recent advances in the field involving signalling and disease, and the involvement of supercomplexes in these mechanisms. Given the importance of mitochondrial ROS, we also provide a schematic guide aimed to help in deciphering the mechanisms involved in their production in a variety of physiological and pathological settings.
Collapse
|
4
|
Saarti M, Almukhtar H, Smith PA, Roberts RE. Effect of mitochondrial complex III inhibitors on the regulation of vascular tone in porcine coronary artery. Eur J Pharmacol 2021; 896:173917. [PMID: 33529727 DOI: 10.1016/j.ejphar.2021.173917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 01/06/2021] [Accepted: 01/26/2021] [Indexed: 11/30/2022]
Abstract
In order to gain insight into the regulation of vascular tone by mitochondria, the effects of mitochondrial complex III inhibitors on contractile responses in porcine isolated coronary arteries were investigated. Segments of porcine coronary arteries were set up for isometric tension recording and concentration response curves to contractile agents were carried out in the absence or presence of the complex III inhibitors antimycin A or myxothiazol. Activity of AMP kinase was determined by measuring changes in phosphorylation of AMP kinase at Thr172. Pre-incubation with 10 μM antimycin A (Qi site inhibitor), or myxothiazol (Qo site inhibitor) led to inhibition of the contraction to the thromboxane receptor agonist U46619. Similar effects were seen on contractile responses to extracellular calcium, and the L-type calcium channel opener BAY K 8644, suggesting that both antimycin A and myxothiazol inhibit calcium-dependent contractions. The inhibitory effect of antimycin A was still seen in the absence of extracellular calcium, indicating an additional effect on a calcium independent pathway. The AMP kinase inhibitor dorsomorphin (10 μM) prevented the inhibitory of antimycin A but not myxothiazol. Furthermore, antimycin A increased the phosphorylation of AMP kinase, indicating an increase in activity, suggesting that antimycin A also acts through this pathway. These data indicate that inhibition of complex III attenuates contractile responses through inhibition of calcium influx. However, inhibition of the Qi site can also inhibit the contractile response through activation of AMP kinase.
Collapse
Affiliation(s)
- Mohammed Saarti
- School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Hani Almukhtar
- School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Paul A Smith
- School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Richard E Roberts
- School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| |
Collapse
|
5
|
AMPK breathing and oxygen supply. Respir Physiol Neurobiol 2019; 265:112-120. [DOI: 10.1016/j.resp.2018.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/06/2018] [Accepted: 08/31/2018] [Indexed: 01/28/2023]
|
6
|
Schleifer G, Marutani E, Ferrari M, Sharma R, Skinner O, Goldberger O, Grange RMH, Peneyra K, Malhotra R, Wepler M, Ichinose F, Bloch DB, Mootha VK, Zapol WM. Impaired hypoxic pulmonary vasoconstriction in a mouse model of Leigh syndrome. Am J Physiol Lung Cell Mol Physiol 2018; 316:L391-L399. [PMID: 30520688 PMCID: PMC6397345 DOI: 10.1152/ajplung.00419.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is a physiological vasomotor response that maintains systemic oxygenation by matching perfusion to ventilation during alveolar hypoxia. Although mitochondria appear to play an essential role in HPV, the impact of mitochondrial dysfunction on HPV remains incompletely defined. Mice lacking the mitochondrial complex I (CI) subunit Ndufs4 ( Ndufs4-/-) develop a fatal progressive encephalopathy and serve as a model for Leigh syndrome, the most common mitochondrial disease in children. Breathing normobaric 11% O2 prevents neurological disease and improves survival in Ndufs4-/- mice. In this study, we found that either genetic Ndufs4 deficiency or pharmacological inhibition of CI using piericidin A impaired the ability of left mainstem bronchus occlusion (LMBO) to induce HPV. In mice breathing air, the partial pressure of arterial oxygen during LMBO was lower in Ndufs4-/- and in piericidin A-treated Ndufs4+/+ mice than in respective controls. Impairment of HPV in Ndufs4-/- mice was not a result of nonspecific dysfunction of the pulmonary vascular contractile apparatus or pulmonary inflammation. In Ndufs4-deficient mice, 3 wk of breathing 11% O2 restored HPV in response to LMBO. When compared with Ndufs4-/- mice breathing air, chronic hypoxia improved systemic oxygenation during LMBO. The results of this study show that, when breathing air, mice with a congenital Ndufs4 deficiency or chemically inhibited CI function have impaired HPV. Our study raises the possibility that patients with inborn errors of mitochondrial function may also have defects in HPV.
Collapse
Affiliation(s)
- Grigorij Schleifer
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Eizo Marutani
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Michele Ferrari
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Rohit Sharma
- Howard Hughes Medical Institute and Department of Molecular Biology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Owen Skinner
- Howard Hughes Medical Institute and Department of Molecular Biology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Olga Goldberger
- Howard Hughes Medical Institute and Department of Molecular Biology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Robert Matthew Henry Grange
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Kathryn Peneyra
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Rajeev Malhotra
- Cardiology Division and Cardiovascular Research Center, Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Martin Wepler
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts.,Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung , Ulm , Germany
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Donald B Bloch
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts.,Division of Rheumatology, Allergy and Immunology, Department of Medicine, Harvard Medical School and Massachusetts General Hospital , Boston, Massachusetts
| | - Vamsi K Mootha
- Howard Hughes Medical Institute and Department of Molecular Biology, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| | - Warren M Zapol
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
7
|
Puukila S, Fernandes RO, Türck P, Carraro CC, Bonetto JHP, de Lima-Seolin BG, da Rosa Araujo AS, Belló-Klein A, Boreham D, Khaper N. Secoisolariciresinol diglucoside attenuates cardiac hypertrophy and oxidative stress in monocrotaline-induced right heart dysfunction. Mol Cell Biochem 2017; 432:33-39. [PMID: 28321539 DOI: 10.1007/s11010-017-2995-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 03/04/2017] [Indexed: 10/19/2022]
Abstract
Pulmonary arterial hypertension (PAH) occurs when remodeling of pulmonary vessels leads to increased pulmonary vascular resistance resulting in increased pulmonary arterial pressure. Increased pulmonary arterial pressure results in right ventricle hypertrophy and eventually heart failure. Oxidative stress has been implicated in the pathogenesis of PAH and may play a role in the regulation of cellular signaling involved in cardiac response to pressure overload. Secoisolariciresinol diglucoside (SDG), a component from flaxseed, has been shown to reduce cardiac oxidative stress in various pathophysiological conditions. We investigated the potential protective effects of SDG in a monocrotaline-induced model of PAH. Five- to six-week-old male Wistar rats were given a single intraperitoneal injection of monocrotaline (60 mg/kg) and sacrificed 21 days later where heart, lung, and plasma were collected. SDG (25 mg/kg) was given via gavage as either a 21-day co-treatment or pre-treatment of 14 days before monocrotaline administration and continued for 21 days. Monocrotaline led to right ventricle hypertrophy, increased lipid peroxidation, and elevated plasma levels of alanine transaminase (ALT) and aspartate transaminase (AST). Co-treatment with SDG did not attenuate hypertrophy or ALT and AST levels but decreased reactive oxygen species (ROS) levels and catalase and superoxide dismutase activity compared to the monocrotaline-treated group. Pre-treatment with SDG decreased right ventricle hypertrophy, ROS levels, lipid peroxidation, catalase, superoxide dismutase, and glutathione peroxidase activity and plasma levels of ALT and AST when compared to the monocrotaline group. These findings indicate that pre-treatment with SDG provided better protection than co-treatment in this model of right heart dysfunction, suggesting an important role for SDG in PAH and right ventricular remodeling.
Collapse
Affiliation(s)
- Stephanie Puukila
- Department of Biology, Lakehead University, 955 Oliver Road, Thunder Bay, ON, P7B 5E1, Canada
| | - Rafael Oliveira Fernandes
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Porto Alegre, RS, 90050-170, Brazil.,Department of Pediatrics, Sainte-Justine University Hospital Research Center, University of Montreal, 3175 Côte Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Patrick Türck
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Porto Alegre, RS, 90050-170, Brazil
| | - Cristina Campos Carraro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Porto Alegre, RS, 90050-170, Brazil
| | - Jéssica Hellen Poletto Bonetto
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Porto Alegre, RS, 90050-170, Brazil
| | - Bruna Gazzi de Lima-Seolin
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Porto Alegre, RS, 90050-170, Brazil
| | - Alex Sander da Rosa Araujo
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Porto Alegre, RS, 90050-170, Brazil
| | - Adriane Belló-Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, Porto Alegre, RS, 90050-170, Brazil
| | - Douglas Boreham
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada
| | - Neelam Khaper
- Medical Sciences Division, Northern Ontario School of Medicine, Lakehead University, 955 Oliver Road, Thunder Bay, ON, P7B 5E1, Canada.
| |
Collapse
|
8
|
Di Mise A, Wang YX, Zheng YM. Role of Transcription Factors in Pulmonary Artery Smooth Muscle Cells: An Important Link to Hypoxic Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:13-32. [PMID: 29047078 DOI: 10.1007/978-3-319-63245-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, namely a lack of oxygen in the blood, induces pulmonary vasoconstriction and vasoremodeling, which serve as essential pathologic factors leading to pulmonary hypertension (PH). The underlying molecular mechanisms are uncertain; however, pulmonary artery smooth muscle cells (PASMCs) play an essential role in hypoxia-induced pulmonary vasoconstriction, vasoremodeling, and PH. Hypoxia causes oxidative damage to DNAs, proteins, and lipids. This damage (oxidative stress) modulates the activity of ion channels and elevates the intracellular calcium concentration ([Ca2+]i, Ca2+ signaling) of PASMCs. The oxidative stress and increased Ca2+ signaling mutually interact with each other, and synergistically results in a variety of cellular responses. These responses include functional and structural abnormalities of mitochondria, sarcoplasmic reticulum, and nucleus; cell contraction, proliferation, migration, and apoptosis, as well as generation of vasoactive substances, inflammatory molecules, and growth factors that mediate the development of PH. A number of studies reveal that various transcription factors (TFs) play important roles in hypoxia-induced oxidative stress, disrupted PAMSC Ca2+ signaling and the development and progress of PH. It is believed that in the pathogenesis of PH, hypoxia facilitates these roles by mediating the expression of multiple genes. Therefore, the identification of specific genes and their transcription factors implicated in PH is necessary for the complete understanding of the underlying molecular mechanisms. Moreover, this identification may aid in the development of novel and effective therapeutic strategies for PH.
Collapse
Affiliation(s)
- Annarita Di Mise
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| | - Yun-Min Zheng
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
9
|
Evans AM, Mahmoud AD, Moral-Sanz J, Hartmann S. The emerging role of AMPK in the regulation of breathing and oxygen supply. Biochem J 2016; 473:2561-72. [PMID: 27574022 PMCID: PMC5003690 DOI: 10.1042/bcj20160002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/20/2016] [Accepted: 05/03/2016] [Indexed: 01/25/2023]
Abstract
Regulation of breathing is critical to our capacity to accommodate deficits in oxygen availability and demand during, for example, sleep and ascent to altitude. It is generally accepted that a fall in arterial oxygen increases afferent discharge from the carotid bodies to the brainstem and thus delivers increased ventilatory drive, which restores oxygen supply and protects against hypoventilation and apnoea. However, the precise molecular mechanisms involved remain unclear. We recently identified as critical to this process the AMP-activated protein kinase (AMPK), which is key to the cell-autonomous regulation of metabolic homoeostasis. This observation is significant for many reasons, not least because recent studies suggest that the gene for the AMPK-α1 catalytic subunit has been subjected to natural selection in high-altitude populations. It would appear, therefore, that evolutionary pressures have led to AMPK being utilized to regulate oxygen delivery and thus energy supply to the body in the short, medium and longer term. Contrary to current consensus, however, our findings suggest that AMPK regulates ventilation at the level of the caudal brainstem, even when afferent input responses from the carotid body are normal. We therefore hypothesize that AMPK integrates local hypoxic stress at defined loci within the brainstem respiratory network with an index of peripheral hypoxic status, namely afferent chemosensory inputs. Allied to this, AMPK is critical to the control of hypoxic pulmonary vasoconstriction and thus ventilation-perfusion matching at the lungs and may also determine oxygen supply to the foetus by, for example, modulating utero-placental blood flow.
Collapse
Affiliation(s)
- A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K.
| | - Amira D Mahmoud
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| | - Javier Moral-Sanz
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| | - Sandy Hartmann
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| |
Collapse
|
10
|
Sommer N, Strielkov I, Pak O, Weissmann N. Oxygen sensing and signal transduction in hypoxic pulmonary vasoconstriction. Eur Respir J 2015; 47:288-303. [PMID: 26493804 DOI: 10.1183/13993003.00945-2015] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/24/2015] [Indexed: 01/17/2023]
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.
Collapse
Affiliation(s)
- Natascha Sommer
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Ievgen Strielkov
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Oleg Pak
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
11
|
Evans AM, Lewis SA, Ogunbayo OA, Moral-Sanz J. Modulation of the LKB1-AMPK Signalling Pathway Underpins Hypoxic Pulmonary Vasoconstriction and Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 860:89-99. [PMID: 26303471 DOI: 10.1007/978-3-319-18440-1_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Perhaps the defining characteristic of pulmonary arteries is the process of hypoxic pulmonary vasoconstriction (HPV) which, under physiological conditions, supports ventilation-perfusion matching in the lung by diverting blood flow away from oxygen deprived areas of the lung to oxygen rich regions. However, when alveolar hypoxia is more widespread, either at altitude or with disease (e.g., cystic fibrosis), HPV may lead to hypoxic pulmonary hypertension. HPV is driven by the intrinsic response to hypoxia of pulmonary arterial smooth muscle and endothelial cells, which are acutely sensitive to relatively small changes in pO2 and have evolved to monitor oxygen supply and thus address ventilation-perfusion mismatch. There is now a consensus that the inhibition by hypoxia of mitochondrial oxidative phosphorylation represents a key step towards the induction of HPV, but the precise nature of the signalling pathway(s) engaged thereafter remains open to debate. We will consider the role of the AMP-activated protein kinase (AMPK) and liver kinase B1 (LKB1), an upstream kinase through which AMPK is intimately coupled to changes in oxygen supply via mitochondrial metabolism. A growing body of evidence, from our laboratory and others, suggests that modulation of the LKB1-AMPK signalling pathway underpins both hypoxic pulmonary vasoconstriction and the development of pulmonary hypertension.
Collapse
Affiliation(s)
- A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK,
| | | | | | | |
Collapse
|
12
|
Strielkov I, Kizub I, Khromov A, Soloviev A. Evidence for the role of phosphatidylcholine-specific phospholipase C in sustained hypoxic pulmonary vasoconstriction. Vascul Pharmacol 2013; 58:292-8. [DOI: 10.1016/j.vph.2013.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/31/2013] [Accepted: 02/04/2013] [Indexed: 11/29/2022]
|
13
|
Waypa GB, Marks JD, Guzy RD, Mungai PT, Schriewer JM, Dokic D, Ball MK, Schumacker PT. Superoxide generated at mitochondrial complex III triggers acute responses to hypoxia in the pulmonary circulation. Am J Respir Crit Care Med 2013; 187:424-32. [PMID: 23328522 DOI: 10.1164/rccm.201207-1294oc] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
RATIONALE The role of reactive oxygen species (ROS) signaling in the O(2) sensing mechanism underlying acute hypoxic pulmonary vasoconstriction (HPV) has been controversial. Although mitochondria are important sources of ROS, studies using chemical inhibitors have yielded conflicting results, whereas cellular models using genetic suppression have precluded in vivo confirmation. Hence, genetic animal models are required to test mechanistic hypotheses. OBJECTIVES We tested whether mitochondrial Complex III is required for the ROS signaling and vasoconstriction responses to acute hypoxia in pulmonary arteries (PA). METHODS A mouse permitting Cre-mediated conditional deletion of the Rieske iron-sulfur protein (RISP) of Complex III was generated. Adenoviral Cre recombinase was used to delete RISP from isolated PA vessels or smooth muscle cells (PASMC). MEASUREMENTS AND MAIN RESULTS In PASMC, RISP depletion abolished hypoxia-induced increases in ROS signaling in the mitochondrial intermembrane space and cytosol, and it abrogated hypoxia-induced increases in [Ca(2+)](i). In isolated PA vessels, RISP depletion abolished hypoxia-induced ROS signaling in the cytosol. Breeding the RISP mice with transgenic mice expressing tamoxifen-activated Cre in smooth muscle permitted the depletion of RISP in PASMC in vivo. Precision-cut lung slices from those mice revealed that RISP depletion abolished hypoxia-induced increases in [Ca(2+)](i) of the PA. In vivo RISP depletion in smooth muscle attenuated the acute hypoxia-induced increase in right ventricular systolic pressure in anesthetized mice. CONCLUSIONS Acute hypoxia induces superoxide release from Complex III of smooth muscle cells. These oxidant signals diffuse into the cytosol and trigger increases in [Ca(2+)](i) that cause acute hypoxic pulmonary vasoconstriction.
Collapse
Affiliation(s)
- Gregory B Waypa
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Tabima DM, Frizzell S, Gladwin MT. Reactive oxygen and nitrogen species in pulmonary hypertension. Free Radic Biol Med 2012; 52:1970-86. [PMID: 22401856 PMCID: PMC3856647 DOI: 10.1016/j.freeradbiomed.2012.02.041] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 02/23/2012] [Accepted: 02/25/2012] [Indexed: 02/07/2023]
Abstract
Pulmonary vascular disease can be defined as either a disease affecting the pulmonary capillaries and pulmonary arterioles, termed pulmonary arterial hypertension, or a disease affecting the left ventricle, called pulmonary venous hypertension. Pulmonary arterial hypertension (PAH) is a disorder of the pulmonary circulation characterized by endothelial dysfunction, as well as intimal and smooth muscle proliferation. Progressive increases in pulmonary vascular resistance and pressure impair the performance of the right ventricle, resulting in declining cardiac output, reduced exercise capacity, right-heart failure, and ultimately death. While the primary and heritable forms of the disease are thought to affect over 5000 patients in the United States, the disease can occur secondary to congenital heart disease, most advanced lung diseases, and many systemic diseases. Multiple studies implicate oxidative stress in the development of PAH. Further, this oxidative stress has been shown to be associated with alterations in reactive oxygen species (ROS), reactive nitrogen species (RNS), and nitric oxide (NO) signaling pathways, whereby bioavailable NO is decreased and ROS and RNS production are increased. Many canonical ROS and NO signaling pathways are simultaneously disrupted in PAH, with increased expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and xanthine oxidoreductase, uncoupling of endothelial NO synthase (eNOS), and reduction in mitochondrial number, as well as impaired mitochondrial function. Upstream dysregulation of ROS/NO redox homeostasis impairs vascular tone and contributes to the pathological activation of antiapoptotic and mitogenic pathways, leading to cell proliferation and obliteration of the vasculature. This paper will review the available data regarding the role of oxidative and nitrosative stress and endothelial dysfunction in the pathophysiology of pulmonary hypertension, and provide a description of targeted therapies for this disease.
Collapse
Affiliation(s)
- Diana M. Tabima
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213
| | - Sheila Frizzell
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213
| | - Mark T. Gladwin
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, 15213
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213
| |
Collapse
|
15
|
Abstract
It has been known for more than 60 years, and suspected for over 100, that alveolar hypoxia causes pulmonary vasoconstriction by means of mechanisms local to the lung. For the last 20 years, it has been clear that the essential sensor, transduction, and effector mechanisms responsible for hypoxic pulmonary vasoconstriction (HPV) reside in the pulmonary arterial smooth muscle cell. The main focus of this review is the cellular and molecular work performed to clarify these intrinsic mechanisms and to determine how they are facilitated and inhibited by the extrinsic influences of other cells. Because the interaction of intrinsic and extrinsic mechanisms is likely to shape expression of HPV in vivo, we relate results obtained in cells to HPV in more intact preparations, such as intact and isolated lungs and isolated pulmonary vessels. Finally, we evaluate evidence regarding the contribution of HPV to the physiological and pathophysiological processes involved in the transition from fetal to neonatal life, pulmonary gas exchange, high-altitude pulmonary edema, and pulmonary hypertension. Although understanding of HPV has advanced significantly, major areas of ignorance and uncertainty await resolution.
Collapse
Affiliation(s)
- J T Sylvester
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, The Johns Hopkins University School ofMedicine, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
16
|
Korde AS, Yadav VR, Zheng YM, Wang YX. Primary role of mitochondrial Rieske iron-sulfur protein in hypoxic ROS production in pulmonary artery myocytes. Free Radic Biol Med 2011; 50:945-52. [PMID: 21238580 PMCID: PMC3051030 DOI: 10.1016/j.freeradbiomed.2011.01.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 01/04/2011] [Accepted: 01/06/2011] [Indexed: 12/20/2022]
Abstract
This study was designed to determine whether: (1) hypoxia could directly affect ROS production in isolated mitochondria and mitochondrial complex III from pulmonary artery smooth muscle cells (PASMCs) and (2) Rieske iron-sulfur protein in complex III might mediate hypoxic ROS production, leading to hypoxic pulmonary vasoconstriction (HPV). Our data, for the first time, demonstrate that hypoxia significantly enhances ROS production, measured by the standard ROS indicator dichlorodihydrofluorescein/diacetate, in isolated mitochondria from PASMCs. Studies using the newly developed, specific ROS biosensor pHyPer have found that hypoxia increases mitochondrial ROS generation in isolated PASMCs as well. Hypoxic ROS production has also been observed in isolated complex III. Rieske iron-sulfur protein silencing using siRNA abolishes the hypoxic ROS formation in isolated PASM complex III, mitochondria, and cells, whereas Rieske iron-sulfur protein overexpression produces the opposite effect. Rieske iron-sulfur protein silencing inhibits the hypoxic increase in [Ca(2+)](i) in PASMCs and hypoxic vasoconstriction in isolated PAs. These findings together provide novel evidence that mitochondria are the direct hypoxic targets in PASMCs, in which Rieske iron-sulfur protein in complex III may serve as an essential, primary molecule that mediates the hypoxic ROS generation, leading to an increase in intracellular Ca(2+) in PASMCs and HPV.
Collapse
Affiliation(s)
| | | | | | - Yong-Xiao Wang
- Corresponding author: Dr. Yong-Xiao Wang Albany Medical College Center for Cardiovascular Sciences (MC-8) 47 New Scotland Avenue Albany, NY 12208 Phone: (518)-262-9506 Fax: (518)-262-8101
| |
Collapse
|
17
|
Evans AM, Hardie DG, Peers C, Mahmoud A. Hypoxic pulmonary vasoconstriction: mechanisms of oxygen-sensing. Curr Opin Anaesthesiol 2011; 24:13-20. [PMID: 21157304 PMCID: PMC3154643 DOI: 10.1097/aco.0b013e3283421201] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Hypoxic pulmonary vasoconstriction (HPV) is driven by the intrinsic response to hypoxia of pulmonary arterial smooth muscle and endothelial cells. These are representatives of a group of specialized O2-sensing cells, defined by their acute sensitivity to relatively small changes in pO2, which have evolved to modulate respiratory and circulatory function in order to maintain O2 supply within physiological limits. The aim of this article is to discuss recent investigations into the mechanism(s) of hypoxia-response coupling and, in light of these, provide a critical assessment of current working hypotheses. RECENT FINDINGS Upon exposure to hypoxia state-of-the-art technologies have now confirmed that mitochondrial oxidative phosphorylation is inhibited in all O2-sensing cells, including pulmonary arterial smooth muscle cells. Thereafter, evidence has been presented to indicate a role as principal effector for the 'gasotransmitters' carbon monoxide and hydrogen sulphide, reactive oxygen species or, in marked contrast, reduced cellular redox couples. Considering recent evidence in favour and against these proposals we suggest that an alternative mechanism may be key, namely the activation of adenosine monophosphate-activated protein kinase consequent to inhibition of mitochondrial oxidative phosphorylation. SUMMARY HPV supports ventilation-perfusion matching in the lung by diverting blood flow away from oxygen-deprived areas towards regions rich in O2. However, in diseases such as emphysema and cystic fibrosis, widespread HPV leads to hypoxic pulmonary hypertension and ultimately right heart failure. Determining the precise mechanism(s) that underpins hypoxia-response coupling will therefore advance understanding of the fundamental processes contributing to related pathophysiology and provide for improved therapeutics.
Collapse
Affiliation(s)
- A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.
| | | | | | | |
Collapse
|
18
|
Chung HH, Dai ZK, Wu BN, Yeh JL, Chai CY, Chu KS, Liu CP, Chen IJ. The xanthine derivative KMUP-1 inhibits models of pulmonary artery hypertension via increased NO and cGMP-dependent inhibition of RhoA/Rho kinase. Br J Pharmacol 2010; 160:971-86. [PMID: 20590592 DOI: 10.1111/j.1476-5381.2010.00740.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE KMUP-1 is known to increase cGMP, enhance endothelial nitric oxide synthase (eNOS) and suppress Rho kinase (ROCK) expression in smooth muscle. Here, we investigated the mechanism of action of KMUP-1 on acute and chronic pulmonary artery hypertension (PAH) in rats. EXPERIMENTAL APPROACH We measured pulmonary vascular contractility, wall thickening, eNOS immunostaining, expressions of ROCK II, RhoA activation, myosin phosphatase target subunit 1 (MYPT1) phosphorylation, eNOS, soluble guanylyl cyclase (sGC), protein kinase G (PKG) and phosphodiesterase 5A (PDE-5A), blood oxygenation and cGMP/cAMP, and right ventricular hypertrophy (RVH) in rats. KEY RESULTS In rings of intact pulmonary artery (PA), KMUP-1 relaxed the vasoconstriction induced by phenylephrine (10 microM) or the thromboxane A(2)-mimetic U46619 (0.5 microM). In endothelium-denuded PA rings, this relaxation was reduced. In acute PAH induced by U46619 (2.5 microg x kg(-1) x min(-1), 30 min), KMUP-1 relaxed vasoconstriction by enhancing levels of eNOS, sGC and PKG, suppressing those of PDE-5A, RhoA/ROCK II activation and MYPT1 phosphorylation, and restoring oxygenation in blood and cGMP/cAMP in plasma. Incubating smooth muscle cells from PA (PASMCs) with KMUP-1 inhibited thapsigargin-induced Ca(2+) efflux and angiotensin II-induced Ca(2+) influx. In chronic PAH model induced by monocrotaline, KMUP-1 increased eNOS and reduced RhoA/ROCK II activation/expression, PA wall thickening, eNOS immunostaining and RVH. KMUP-1 and sildenafil did not inhibit monocrotaline-induced PDE-5A expression. CONCLUSION AND IMPLICATIONS KMUP-1 decreased PAH by enhancing NO synthesis by eNOS, with consequent cGMP-dependent inhibition of RhoA/ROCK II and Ca(2+) desensitization in PASMCs. KMUP-1 has the potential to reduce vascular resistance, remodelling and RVH in PAH.
Collapse
Affiliation(s)
- Hui-Hsuan Chung
- Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Fuchs B, Sommer N, Dietrich A, Schermuly RT, Ghofrani HA, Grimminger F, Seeger W, Gudermann T, Weissmann N. Redox signaling and reactive oxygen species in hypoxic pulmonary vasoconstriction. Respir Physiol Neurobiol 2010; 174:282-91. [PMID: 20801235 DOI: 10.1016/j.resp.2010.08.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 08/20/2010] [Accepted: 08/20/2010] [Indexed: 11/16/2022]
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is an essential physiological mechanism of the lung that matches blood perfusion with alveolar ventilation to optimize gas exchange. Perturbations of HPV, as may occur in pneumonia or adult respiratory distress syndrome, can cause life-threatening hypoxemia. Despite intensive research for decades, the molecular mechanisms of HPV have not been fully elucidated. Reactive oxygen species (ROS) and changes in the cellular redox state are proposed to link O2 sensing and pulmonary arterial smooth muscle cell contraction underlying HPV. In this regard, mitochondria and NAD(P)H oxidases are discussed as sources of ROS. However, there is controversy whether ROS levels decrease or increase during hypoxia. With this background we summarize the current knowledge on the role of ROS and redox state in HPV.
Collapse
Affiliation(s)
- Beate Fuchs
- Excellence Cluster Cardio-Pulmonary System, University of Giessen Lung Center, Department of Internal Medicine II, Justus-Liebig-University Giessen, Aulweg 130, 35392 Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Interactions between calcium and reactive oxygen species in pulmonary arterial smooth muscle responses to hypoxia. Respir Physiol Neurobiol 2010; 174:221-9. [PMID: 20801238 DOI: 10.1016/j.resp.2010.08.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 08/18/2010] [Accepted: 08/20/2010] [Indexed: 02/07/2023]
Abstract
In contrast to the systemic vasculature, where hypoxia causes vasodilation, pulmonary arteries constrict in response to hypoxia. The mechanisms underlying this unique response have been the subject of investigation for over 50 years, and still remain a topic of great debate. Over the last 20 years, there has emerged a general consensus that both increases in intracellular calcium concentration and changes in reactive oxygen species (ROS) generation play key roles in the pulmonary vascular response to hypoxia. Controversy exists, however, regarding whether ROS increase or decrease during hypoxia, the source of ROS, and the mechanisms by which changes in ROS might impact intracellular calcium, and vice versa. This review will discuss the mechanisms regulating [Ca2+]i and ROS in PASMCs, and the interaction between ROS and Ca2+ signaling during exposure to acute hypoxia.
Collapse
|
21
|
Wang YX, Zheng YM. Role of ROS signaling in differential hypoxic Ca2+ and contractile responses in pulmonary and systemic vascular smooth muscle cells. Respir Physiol Neurobiol 2010; 174:192-200. [PMID: 20713188 DOI: 10.1016/j.resp.2010.08.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Revised: 08/06/2010] [Accepted: 08/09/2010] [Indexed: 01/25/2023]
Abstract
Hypoxia causes a large increase in [Ca2+]i and attendant contraction in pulmonary artery smooth muscle cells (PASMCs), but not in systemic artery SMCs. The different responses meet the respective functional needs in these two distinct vascular myocytes; however, the underlying molecular mechanisms are not well known. We and other investigators have provided extensive evidence to reveal that voltage-dependent K+ (KV) channels, canonical transient receptor potential (TRPC) channels, ryanodine receptor Ca2+ release channels (RyRs), cyclic adenosine diphosphate-ribose, FK506 binding protein 12.6, protein kinase C, NADPH oxidase and reactive oxygen species (ROS) are the essential effectors and signaling intermediates in the hypoxic increase in [Ca2+]i in PASMCs and HPV, but they may not primarily underlie the diverse cellular responses in pulmonary and systemic vascular myocytes. Hypoxia significantly increases mitochondrial ROS generation in PASMCs, which can induce intracellular Ca2+ release by opening RyRs, and may also cause extracellular Ca2+ influx by inhibiting KV channels and activating TRPC channels, leading to a large increase in [Ca2+]i in PASMCs and HPV. In contrast, hypoxia has no or a minor effect on mitochondrial ROS generation in systemic SMCs, thereby causing no change or a negligible increase in [Ca2+]i and contraction. Further preliminary work indicates that Rieske iron-sulfur protein in the mitochondrial complex III may perhaps serve as a key initial molecular determinant for the hypoxic increase in [Ca2+]i in PASMCs and HPV, suggesting its potential important role in different cellular changes to respond to hypoxic stimulation in pulmonary and systemic artery myocytes. All these findings have greatly improved our understanding of the molecular processes for the differential hypoxic Ca2+ and contractile responses in vascular SMCs from distinct pulmonary and systemic circulation systems.
Collapse
Affiliation(s)
- Yong-Xiao Wang
- Center for Cardiovascular Sciences, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA.
| | | |
Collapse
|
22
|
Waypa GB, Schumacker PT. Hypoxia-induced changes in pulmonary and systemic vascular resistance: where is the O2 sensor? Respir Physiol Neurobiol 2010; 174:201-11. [PMID: 20713189 DOI: 10.1016/j.resp.2010.08.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 08/05/2010] [Accepted: 08/06/2010] [Indexed: 01/06/2023]
Abstract
Pulmonary arteries (PA) constrict in response to alveolar hypoxia, whereas systemic arteries (SA) undergo dilation. These physiological responses reflect the need to improve gas exchange in the lung, and to enhance the delivery of blood to hypoxic systemic tissues. An important unresolved question relates to the underlying mechanism by which the vascular cells detect a decrease in oxygen tension and translate that into a signal that triggers the functional response. A growing body of work implicates the mitochondria, which appear to function as O2 sensors by initiating a redox-signaling pathway that leads to the activation of downstream effectors that regulate vascular tone. However, the direction of this redox signal has been the subject of controversy. Part of the problem has been the lack of appropriate tools to assess redox signaling in live cells. Recent advancements in the development of redox sensors have led to studies that help to clarify the nature of the hypoxia-induced redox signaling by reactive oxygen species (ROS). Moreover, these studies provide valuable insight regarding the basis for discrepancies in earlier studies of the hypoxia-induced mechanism of redox signaling. Based on recent work, it appears that the O2 sensing mechanism in both the PA and SA are identical, that mitochondria function as the site of O2 sensing, and that increased ROS release from these organelles leads to the activation of cell-specific, downstream vascular responses.
Collapse
Affiliation(s)
- Gregory B Waypa
- Department of Pediatrics, Division of Neonatology, Northwestern University, Morton Building 4-685, 310 East Superior St, Chicago, IL 60611, USA.
| | | |
Collapse
|
23
|
Chandel NS. Mitochondrial complex III: an essential component of universal oxygen sensing machinery? Respir Physiol Neurobiol 2010; 174:175-81. [PMID: 20708106 DOI: 10.1016/j.resp.2010.08.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 08/03/2010] [Accepted: 08/04/2010] [Indexed: 02/07/2023]
Abstract
Oxygen is necessary for the survival of mammalian cells. In order to maintain adequate cellular oxygenation, mammals have evolved multiple acute and long-term adaptive responses to hypoxia. These include hypoxic increases in erythropoiesis, pulmonary vasoconstriction and carotid body neurosecretion. Collectively, these responses help maintain oxygen homeostasis as oxygen levels remain scarce. There are multiple effectors proposed to underlie these diverse responses to hypoxia including PHD2, AMPK, NADPH oxidases, and mitochondrial complex III. Here I propose a model wherein complex III is integral to oxygen sensing in regulating diverse response to hypoxia.
Collapse
Affiliation(s)
- Navdeep S Chandel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Medical School, Chicago, IL 60611, USA.
| |
Collapse
|
24
|
Wang YX, Zheng YM. ROS-dependent signaling mechanisms for hypoxic Ca(2+) responses in pulmonary artery myocytes. Antioxid Redox Signal 2010; 12:611-23. [PMID: 19764882 PMCID: PMC2861542 DOI: 10.1089/ars.2009.2877] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Hypoxic exposure causes pulmonary vasoconstriction, which serves as a critical physiologic process that ensures regional alveolar ventilation and pulmonary perfusion in the lungs, but may become an essential pathologic factor leading to pulmonary hypertension. Although the molecular mechanisms underlying hypoxic pulmonary vasoconstriction and associated pulmonary hypertension are uncertain, increasing evidence indicates that hypoxia can result in a significant increase in intracellular reactive oxygen species concentration ([ROS](i)) through the mitochondrial electron-transport chain in pulmonary artery smooth muscle cells (PASMCs). The increased mitochondrial ROS subsequently activate protein kinase C-epsilon (PKCepsilon) and NADPH oxidase (Nox), providing positive mechanisms that further increase [ROS](i). ROS may directly cause extracellular Ca(2+) influx by inhibiting voltage-dependent K(+) (K(V)) channels and opening of store-operated Ca(2+) (SOC) channels, as well as intracellular Ca(2+) release by activating ryanodine receptors (RyRs), leading to an increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) and associated contraction. In concert with ROS, PKCepsilon may also affect K(V) channels, SOC channels, and RyRs, contributing to hypoxic Ca(2+) and contractile responses in PASMCs.
Collapse
Affiliation(s)
- Yong-Xiao Wang
- Center for Cardiovascular Sciences, Albany Medical College, New York 12208, USA.
| | | |
Collapse
|
25
|
Zoer B, Cogolludo AL, Perez-Vizcaino F, De Mey JGR, Blanco CE, Villamor E. Hypoxia sensing in the fetal chicken femoral artery is mediated by the mitochondrial electron transport chain. Am J Physiol Regul Integr Comp Physiol 2010; 298:R1026-34. [PMID: 20089711 DOI: 10.1152/ajpregu.00500.2009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vascular hypoxia sensing is transduced into vasoconstriction in the pulmonary circulation, whereas systemic arteries dilate. Mitochondrial electron transport chain (mETC), reactive O(2) species (ROS), and K(+) channels have been implicated in the sensing/signaling mechanisms of hypoxic relaxation in mammalian systemic arteries. We aimed to investigate their putative roles in hypoxia-induced relaxation in fetal chicken (19 days of incubation) femoral arteries mounted in a wire myograph. Acute hypoxia (Po(2) approximately 2.5 kPa) relaxed the contraction induced by norepinephrine (1 microM). Hypoxia-induced relaxation was abolished or significantly reduced by the mETC inhibitors rotenone (complex I), myxothiazol and antimycin A (complex III), and NaN(3) (complex IV). The complex II inhibitor 3-nitroproprionic acid enhanced the hypoxic relaxation. In contrast, the relaxations mediated by acetylcholine, sodium nitroprusside, or forskolin were not affected by the mETC blockers. Hypoxia induced a slight increase in ROS production (as measured by 2,7-dichlorofluorescein-fluorescence), but hypoxia-induced relaxation was not affected by scavenging of superoxide (polyethylene glycol-superoxide dismutase) or H(2)O(2) (polyethylene glycol-catalase) or by NADPH-oxidase inhibition (apocynin). Also, the K(+) channel inhibitors tetraethylammonium (nonselective), diphenyl phosphine oxide-1 (voltage-gated K(+) channel 1.5), glibenclamide (ATP-sensitive K(+) channel), iberiotoxin (large-conductance Ca(2+)-activated K(+) channel), and BaCl(2) (inward-rectifying K(+) channel), as well as ouabain (Na(+)-K(+)-ATPase inhibitor) did not affect hypoxia-induced relaxation. The relaxation was enhanced in the presence of the voltage-gated K(+) channel blocker 4-aminopyridine. In conclusion, our experiments suggest that the mETC plays a critical role in O(2) sensing in fetal chicken femoral arteries. In contrast, hypoxia-induced relaxation appears not to be mediated by ROS or K(+) channels.
Collapse
Affiliation(s)
- Bea Zoer
- University Hospital Maastricht, P. Debyelaan 25, AZ Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
26
|
Waypa GB, Marks JD, Guzy R, Mungai PT, Schriewer J, Dokic D, Schumacker PT. Hypoxia triggers subcellular compartmental redox signaling in vascular smooth muscle cells. Circ Res 2009; 106:526-35. [PMID: 20019331 DOI: 10.1161/circresaha.109.206334] [Citation(s) in RCA: 285] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RATIONALE Recent studies have implicated mitochondrial reactive oxygen species (ROS) in regulating hypoxic pulmonary vasoconstriction (HPV), but controversy exists regarding whether hypoxia increases or decreases ROS generation. OBJECTIVE This study tested the hypothesis that hypoxia induces redox changes that differ among subcellular compartments in pulmonary (PASMCs) and systemic (SASMCs) smooth muscle cells. METHODS AND RESULTS We used a novel, redox-sensitive, ratiometric fluorescent protein sensor (RoGFP) to assess the effects of hypoxia on redox signaling in cultured PASMCs and SASMCs. Using genetic targeting sequences, RoGFP was expressed in the cytosol (Cyto-RoGFP), the mitochondrial matrix (Mito-RoGFP), or the mitochondrial intermembrane space (IMS-RoGFP), allowing assessment of oxidant signaling in distinct intracellular compartments. Superfusion of PASMCs or SASMCs with hypoxic media increased oxidation of both Cyto-RoGFP and IMS-RoGFP. However, hypoxia decreased oxidation of Mito-RoGFP in both cell types. The hypoxia-induced oxidation of Cyto-RoGFP was attenuated through the overexpression of cytosolic catalase in PASMCs. CONCLUSIONS These results indicate that hypoxia causes a decrease in nonspecific ROS generation in the matrix compartment, whereas it increases regulated ROS production in the IMS, which diffuses to the cytosol of both PASMCs and SASMCs.
Collapse
Affiliation(s)
- Gregory B Waypa
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Evans AM, Hardie DG, Peers C, Wyatt CN, Viollet B, Kumar P, Dallas ML, Ross F, Ikematsu N, Jordan HL, Barr BL, Rafferty JN, Ogunbayo O. Ion channel regulation by AMPK: the route of hypoxia-response coupling in thecarotid body and pulmonary artery. Ann N Y Acad Sci 2009; 1177:89-100. [PMID: 19845611 DOI: 10.1111/j.1749-6632.2009.05041.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vital homeostatic mechanisms monitor O2 supply and adjust respiratory and circulatory function to meet demand. The pulmonary arteries and carotid bodies are key systems in this respect. Hypoxic pulmonary vasoconstriction (HPV) aids ventilation-perfusion matching in the lung by diverting blood flow from areas with an O2 deficit to those rich in O2, while a fall in arterial pO2 increases sensory afferent discharge from the carotid body to elicit corrective changes in breathing patterns. We discuss here the new concept that hypoxia, by inhibiting oxidative phosphorylation, activates AMP-activated protein kinase (AMPK) leading to consequent phosphorylation of target proteins, such as ion channels, which initiate pulmonary artery constriction and carotid body activation. Consistent with this view, AMPK knockout mice exhibit an impaired ventilatory response to hypoxia. Thus, AMPK may be sufficient and necessary for hypoxia-response coupling and may regulate O2 and thereby energy (ATP) supply at the whole body as well as the cellular level.
Collapse
Affiliation(s)
- A Mark Evans
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Pullamsetti SS, Schermuly RT. Endothelin receptor antagonists in preclinical models of pulmonary hypertension. Eur J Clin Invest 2009; 39 Suppl 2:3-13. [PMID: 19335741 DOI: 10.1111/j.1365-2362.2009.02115.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Pulmonary hypertension (PH), a chronic disorder of the pulmonary vasculature, is characterized by progressive elevation in pulmonary artery pressure and the ultimate development of right-sided heart failure and death. Being a rapidly progressive disease with limited therapeutic options, the pathogenesis of PH is complex and multifactorial. The pathogenesis may result from a combination of vasoconstriction, inward vascular wall remodelling and in situ thrombosis that involves dysfunction of underlying cellular pathways and mediators. Among these, the activation of endothelin (ET) system has been shown to be important in the development and perpetuation of PH. Endothelin-1 (ET-1), a potent vasoconstrictor and mitogen, exerts its biological effects by binding to two G-protein-coupled receptor isoforms, endothelin A (ETA) receptor and endothelin B (ETB) receptor. These two receptors are nonredundant and unique because of distinct localization, unique binding locations and affinities for the endothelin peptide and activation of distinct signalling pathways. Importantly, there is now substantial evidence that direct antagonism of ET receptors that can block either ETA- or ETA- and ETB receptors can be beneficial for the treatment of PH in both preclinical and clinical setting. This review provides an overview of endothelin biology, various preclinical models that have been widely used to investigate the pathophysiology of PH as well as the individual roles of the ET receptors (ETA and ETB) and their regulation in disease pathogenesis. We also review current data on the use of selective and nonselective ET receptor antagonism in the preclinical PH models.
Collapse
|
29
|
Stowe DF, Camara AKS. Mitochondrial reactive oxygen species production in excitable cells: modulators of mitochondrial and cell function. Antioxid Redox Signal 2009; 11:1373-414. [PMID: 19187004 PMCID: PMC2842133 DOI: 10.1089/ars.2008.2331] [Citation(s) in RCA: 351] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 01/12/2009] [Accepted: 01/13/2009] [Indexed: 12/14/2022]
Abstract
The mitochondrion is a major source of reactive oxygen species (ROS). Superoxide (O(2)(*-)) is generated under specific bioenergetic conditions at several sites within the electron-transport system; most is converted to H(2)O(2) inside and outside the mitochondrial matrix by superoxide dismutases. H(2)O(2) is a major chemical messenger that, in low amounts and with its products, physiologically modulates cell function. The redox state and ROS scavengers largely control the emission (generation scavenging) of O(2)(*-). Cell ischemia, hypoxia, or toxins can result in excess O(2)(*-) production when the redox state is altered and the ROS scavenger systems are overwhelmed. Too much H(2)O(2) can combine with Fe(2+) complexes to form reactive ferryl species (e.g., Fe(IV) = O(*)). In the presence of nitric oxide (NO(*)), O(2)(*-) forms the reactant peroxynitrite (ONOO(-)), and ONOOH-induced nitrosylation of proteins, DNA, and lipids can modify their structure and function. An initial increase in ROS can cause an even greater increase in ROS and allow excess mitochondrial Ca(2+) entry, both of which are factors that induce cell apoptosis and necrosis. Approaches to reduce excess O(2)(*-) emission include selectively boosting the antioxidant capacity, uncoupling of oxidative phosphorylation to reduce generation of O(2)(*-) by inducing proton leak, and reversibly inhibiting electron transport. Mitochondrial cation channels and exchangers function to maintain matrix homeostasis and likely play a role in modulating mitochondrial function, in part by regulating O(2)(*-) generation. Cell-signaling pathways induced physiologically by ROS include effects on thiol groups and disulfide linkages to modify posttranslationally protein structure to activate/inactivate specific kinase/phosphatase pathways. Hypoxia-inducible factors that stimulate a cascade of gene transcription may be mediated physiologically by ROS. Our knowledge of the role played by ROS and their scavenging systems in modulation of cell function and cell death has grown exponentially over the past few years, but we are still limited in how to apply this knowledge to develop its full therapeutic potential.
Collapse
Affiliation(s)
- David F Stowe
- Anesthesiology Research Laboratories, Department of Anesthesiology, The Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | |
Collapse
|
30
|
Rathore R, Zheng YM, Niu CF, Liu QH, Korde A, Ho YS, Wang YX. Hypoxia activates NADPH oxidase to increase [ROS]i and [Ca2+]i through the mitochondrial ROS-PKCepsilon signaling axis in pulmonary artery smooth muscle cells. Free Radic Biol Med 2008; 45:1223-31. [PMID: 18638544 PMCID: PMC2586914 DOI: 10.1016/j.freeradbiomed.2008.06.012] [Citation(s) in RCA: 228] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Accepted: 06/11/2008] [Indexed: 11/23/2022]
Abstract
The importance of NADPH oxidase (Nox) in hypoxic responses in hypoxia-sensing cells, including pulmonary artery smooth muscle cells (PASMCs), remains uncertain. In this study, using Western blot analysis we found that the major Nox subunits Nox1, Nox4, p22(phox), p47(phox), and p67(phox) were equivalently expressed in mouse pulmonary and systemic (mesenteric) arteries. However, acute hypoxia significantly increased Nox activity and translocation of p47(phox) protein to the plasma membrane in pulmonary, but not mesenteric, arteries. The Nox inhibitor apocynin and p47(phox) gene deletion attenuated the hypoxic increase in intracellular concentrations of reactive oxygen species and Ca(2+) ([ROS](i) and [Ca(2+)](i)), as well as contractions in mouse PASMCs, and abolished the hypoxic activation of Nox in pulmonary arteries. The conventional/novel protein kinase C (PKC) inhibitor chelerythrine, specific PKCepsilon translocation peptide inhibitor, and PKCepsilon gene deletion, but not the conventional PKC inhibitor GO6976, prevented the hypoxic increase in Nox activity in pulmonary arteries and [ROS](i) in PASMCs. The PKC activator phorbol 12-myristate 13-acetate could increase Nox activity in pulmonary and mesenteric arteries. Inhibition of mitochondrial ROS generation with rotenone or myxothiazol prevented hypoxic activation of Nox. Glutathione peroxidase-1 (Gpx1) gene overexpression to enhance H(2)O(2) removal significantly inhibited the hypoxic activation of Nox, whereas Gpx1 gene deletion had the opposite effect. Exogenous H(2)O(2) increased Nox activity in pulmonary and mesenteric arteries. These findings suggest that acute hypoxia may distinctively activate Nox to increase [ROS](i) through the mitochondrial ROS-PKCepsilon signaling axis, providing a positive feedback mechanism to contribute to the hypoxic increase in [ROS](i) and [Ca(2+)](i) as well as contraction in PASMCs.
Collapse
Affiliation(s)
- Rakesh Rathore
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
| | - Yun-Min Zheng
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
| | - Chun-Feng Niu
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
| | - Qing-Hua Liu
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
| | - Amit Korde
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
| | - Ye-Shih Ho
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
| | - Yong-Xiao Wang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
- Corresponding author: Dr. Yong-Xiao Wang, Albany Medical College, Center for Cardiovascular Sciences, Albany, NY 12208, Tel: 518 262-6504, Fax: 518 262-8101,
| |
Collapse
|
31
|
Kiss L, Röder Y, Bier J, Weissmann N, Seeger W, Grimminger F. Direct eicosanoid profiling of the hypoxic lung by comprehensive analysis via capillary liquid chromatography with dual online photodiode-array and tandem mass-spectrometric detection. Anal Bioanal Chem 2007; 390:697-714. [PMID: 18080119 DOI: 10.1007/s00216-007-1718-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Revised: 10/12/2007] [Accepted: 10/23/2007] [Indexed: 11/24/2022]
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.
Collapse
Affiliation(s)
- Ladislau Kiss
- Biochemie Med. II, University of Giessen Lung Center, Department of Internal Medicine, Justus Liebig University, Paul Meimberg Str. 5, 35392, Giessen, Germany.
| | | | | | | | | | | |
Collapse
|
32
|
Waypa GB, Schumacker PT. Oxygen sensing in hypoxic pulmonary vasoconstriction: using new tools to answer an age-old question. Exp Physiol 2007; 93:133-8. [PMID: 17993507 DOI: 10.1113/expphysiol.2007.041236] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Hypoxic pulmonary vasoconstriction (HPV) becomes activated in response to alveolar hypoxia and, although the characteristics of HPV have been well described, the underlying mechanism of O(2) sensing which initiates the HPV response has not been fully established. Mitochondria have long been considered as a putative site of oxygen sensing because they consume O(2) and therefore represent the intracellular site with the lowest oxygen tension. However, two opposing theories have emerged regarding mitochondria-dependent O(2) sensing during hypoxia. One model suggests that there is a decrease in mitochondrial reactive oxygen species (ROS) levels during the transition from normoxia to hypoxia, resulting in the shift in cytosolic redox to a more reduced state. An alternative model proposes that hypoxia paradoxically increases mitochondrial ROS signalling in pulmonary arterial smooth muscle. Experimental resolution of the question of whether the mitochondrial ROS levels increase or decrease during hypoxia has been problematic owing to the technical limitations of the tools used to assess oxidant stress as well as the pharmacological agents used to inhibit the mitochondrial electron transport chain. However, recent developments in genetic techniques and redox-sensitive probes may allow us eventually to reach a consensus concerning the O(2) sensing mechanism underlying HPV.
Collapse
Affiliation(s)
- Gregory B Waypa
- Department of Pediatrics, North-western University, Ward Building 12-191, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | | |
Collapse
|
33
|
Miller FJ, Filali M, Huss GJ, Stanic B, Chamseddine A, Barna TJ, Lamb FS. Cytokine activation of nuclear factor kappa B in vascular smooth muscle cells requires signaling endosomes containing Nox1 and ClC-3. Circ Res 2007; 101:663-71. [PMID: 17673675 DOI: 10.1161/circresaha.107.151076] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Reactive oxygen species (ROS) are mediators of intracellular signals for a myriad of normal and pathologic cellular events, including differentiation, hypertrophy, proliferation, and apoptosis. NADPH oxidases are important sources of ROS that are present in diverse tissues throughout the body and activate many redox-sensitive signal transduction and gene expression pathways. To avoid toxicity and provide specificity of signaling, ROS production and metabolism necessitate tight regulation that likely includes subcellular compartmentalization. However, the constituent elements of NADPH oxidase-dependent cell signaling are not known. To address this issue, we examined cytokine generation of ROS and subsequent activation of the transcription factor nuclear factor kappaB in vascular smooth muscle cells (SMCs). Tumor necrosis factor-alpha and interleukin (IL)-1beta stimulation of SMCs resulted in diphenylene iodonium-sensitive ROS production within intracellular vesicles. Nox1 and p22(phox), integral membrane subunits of NADPH oxidase, coimmunoprecipitated with early endosomal markers in SMCs. ClC-3, an anion transporter that is primarily found in intracellular vesicles, also colocalized with Nox1 in early endosomes and was necessary for tumor necrosis factor-alpha and interleukin-1beta generation of ROS. Cytokine activation of nuclear factor kappaB in SMCs required both Nox1 and ClC-3. We conclude that in response to tumor necrosis factor-alpha and interleukin-1beta, NADPH oxidase generates ROS within early endosomes and that Nox1 cannot produce sufficient ROS for cell signaling in the absence of ClC-3. These data best support a model whereby ClC-3 is required for charge neutralization of the electron flow generated by Nox1 across the membrane of signaling endosomes.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Chloride Channels/genetics
- Chloride Channels/physiology
- Cytokines/physiology
- Endosomes/genetics
- Endosomes/metabolism
- Endosomes/physiology
- Humans
- Mice
- Mice, Knockout
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- NADPH Oxidase 1
- NADPH Oxidases/genetics
- NADPH Oxidases/physiology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Signal Transduction/physiology
Collapse
|
34
|
Panee J, Liu W, Nakamura K, Berry MJ. The responses of HT22 cells to the blockade of mitochondrial complexes and potential protective effect of selenium supplementation. Int J Biol Sci 2007; 3:335-41. [PMID: 17657281 PMCID: PMC1925139 DOI: 10.7150/ijbs.3.335] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Accepted: 07/11/2007] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are the major reactive oxygen species (ROS) – generating sites in mammalian cells. Blockade of complexes in the electron transport chain (ETC) increases the leakage of single electrons to O2 and therefore increases ROS levels. Complexes I and III have been reported to be the major ROS-generating sites in mitochondria. In this study, using mouse hippocampal HT22 cells as in vitro model, we monitored the change of intracellular ROS level in response to the blockade of ETC at different complex, and measured changes of gene expression of antioxidant enzymes and phase II enzymes, also evaluated potential protective effect of selenium (Se) supplementation to the cells under this oxidative stress. In summary, our results showed that complex I was the major ROS-generating site in HT22 cells. Complex I blockade upregulated the mRNA levels of glutamylcysteine synthetase heavy and light chains, glutathione-S-transferases omega1 and alpha 2, hemoxygenase 1, thioredoxin reductase 1, and selenoprotein H. Unexpectedly, the expression of the enzymes that directly scavenge ROS decreased, including superoxide dismutases 1 and 2, glutathione peroxidase 1, and catalase. Se supplementation increased glutathione levels and glutathione peroxidase activity, indicating a potential protective role in oxidative stress caused by ETC blockade.
Collapse
Affiliation(s)
- Jun Panee
- 1. Department of Cell & Molecular Biology, John A Burns Medical School, University of Hawaii, Honolulu HI 96813, USA
| | - Wanyu Liu
- 1. Department of Cell & Molecular Biology, John A Burns Medical School, University of Hawaii, Honolulu HI 96813, USA
| | - Kyoko Nakamura
- 2. School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Marla J. Berry
- 1. Department of Cell & Molecular Biology, John A Burns Medical School, University of Hawaii, Honolulu HI 96813, USA
| |
Collapse
|
35
|
Wang QS, Zheng YM, Dong L, Ho YS, Guo Z, Wang YX. Role of mitochondrial reactive oxygen species in hypoxia-dependent increase in intracellular calcium in pulmonary artery myocytes. Free Radic Biol Med 2007; 42:642-53. [PMID: 17291988 PMCID: PMC1852485 DOI: 10.1016/j.freeradbiomed.2006.12.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Revised: 11/29/2006] [Accepted: 12/07/2006] [Indexed: 10/23/2022]
Abstract
Previous studies examining the role of mitochondria-derived reactive oxygen species (ROS) in hypoxic responses have been mainly conducted in isolated lungs and cultured pulmonary artery smooth muscle cells (PASMCs) using mitochondrial inhibitors, and yielded largely conflicting results. Here we report that in freshly isolated mouse PASMCs, which are devoid of the mixed responses from multi-types of cells in lungs and significant changes in gene expression in cultured cells, the mitochondrial electron transport chain (ETC) complex I, II, or III inhibitors blocked hypoxia-induced increases in intracellular ROS and Ca2+ concentration ([ROS]i and [Ca2+]i) without effects on their resting levels. Inhibition of the complex I plus II and/or III did not produce an additive effect. Glutathione peroxidase-1 (Gpx1) or catalase gene overexpression to enhance H2O2 removal remarkably reduced hypoxic increases in [ROS]i and [Ca2+]i, whereas Gpx1 gene deletion had the opposite effect. None of these genetic modifications changed the resting [ROS]i and [Ca2+]i. H2O2 at 51 microM caused a similar increase in DCF fluorescence ([ROS]i) as that by hypoxia, but only induced 33% of hypoxic increase in [Ca2+]i. Moreover, H2O2 (5.1 microM) reversed the inhibition of the hypoxia-induced increase in [Ca2+]i by rotenone. Collectively, our study using various mitochondrial inhibitors and genetic approaches demonstrates that in response to acute hypoxia, the mitochondrial ETC molecules prior to the complex III ubisemiquinone site act as a functional unit to increase the generation of ROS, particularly H2O2, which is important for, but may not fully cause, the hypoxic increase in [Ca2+]i in freshly isolated PASMCs.
Collapse
Affiliation(s)
- Qing-Song Wang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
| | - Yun-Min Zheng
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
| | - Ling Dong
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
| | - Ye-Shih Ho
- Department of Pathology, Anatomy & Cell Biology, Meharry Medical College, Nashville, TN 37208
| | - Zhongmao Guo
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
| | - Yong-Xiao Wang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208
- *Corresponding author: Dr. Yong-Xiao Wang, Albany Medical College, Center for Cardiovascular Sciences, Albany, NY 12208, Tel: 518 262-6504, Fax: 518 262-8101,
| |
Collapse
|
36
|
Hübler M, Heller AR, Bleyl JU, Rössel T, Stehr SN, Koch T. SPATIAL PULMONARY FLOW DISTRIBUTION IN RABBIT ISOLATED LUNGS IS A POOR REPRESENTATION OF THE SITUATION IN VIVO*. Clin Exp Pharmacol Physiol 2007; 34:269-73. [PMID: 17324136 DOI: 10.1111/j.1440-1681.2007.04557.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Isolated lung preparations are established to investigate effects on pulmonary vascular tone and spatial pulmonary flow (Q (rel)) distribution. In the present study, we hypothesized that Q (rel) distribution in isolated lungs is only poorly correlated with the in vivo situation. 2. Fourteen rabbits were anaesthetized and mechanically ventilated with room air. Animals were held in an upright position for 15 min and Q (rel) was assessed using fluorescent microspheres (Q (rel-in vivo)). A second injection of microspheres was made after isolation of the lungs (Q (rel-ex vivo)). Lungs were dried, cut into 1 cm(3) cubes and spatial Q (rel) distributions were analysed. 3. The mean correlation of Q (rel-in vivo) and Q (rel-ex vivo) was 0.592 +/- 0.188 (95% confidence interval 0.493-0.690). The Q (rel) was redistributed to more ventral (the mean slope of Q (rel) vs the dorsal-ventral axis changed from -0.289 +/- 0.227 to -0.147 +/- 0.114; P = 0.03), cranial (mean slope of Q (rel) vs the caudal-cranial axis changed from -0.386 +/- 0.193 to -0.176 +/- 0.142; P < 0.001) and central (mean slope of Q (rel) vs the hilus-peripheral axis changed from 0.436 +/- 0.133 to -0.236 +/- 0.159; P = 0.003) lung areas. 4. The results obtained from studies investigating Q (rel) distributions in isolated lung models must be interpreted cautiously because the isolated lung set-up significantly affects the spatial distribution of pulmonary flow.
Collapse
Affiliation(s)
- M Hübler
- Department of Anaesthesiology and Intensive Care Medicine, Technical University Dresden, Carl Gustav Carus University Hospital, Fetscherstrasse 74, 01307 Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
37
|
Rathore R, Zheng YM, Li XQ, Wang QS, Liu QH, Ginnan RR, Singer HA, Ho YS, Wang YX. Mitochondrial ROS-PKCepsilon signaling axis is uniquely involved in hypoxic increase in [Ca2+]i in pulmonary artery smooth muscle cells. Biochem Biophys Res Commun 2006; 351:784-90. [PMID: 17087917 PMCID: PMC1764638 DOI: 10.1016/j.bbrc.2006.10.116] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Accepted: 10/24/2006] [Indexed: 11/22/2022]
Abstract
The molecular mechanisms underlying hypoxic responses in pulmonary and systemic arteries remain obscure. Here we for the first time report that acute hypoxia significantly increased total PKC and PKCepsilon activity in pulmonary, but not mesenteric arteries, while these two tissues showed comparable PKCepsilon protein expression and activation by the PKC activator phorbol 12-myristate 13-acetate. Hypoxia induced an increase in intracellular reactive oxygen species (ROS) generation in isolated pulmonary artery smooth muscle cells (PASMCs), but not in mesenteric artery SMCs. Inhibition of mitochondrial ROS generation with rotenone, myxothiazol, or glutathione peroxidase-1 overexpression prevented hypoxia-induced increases in total PKC and PKCepsilon activity in pulmonary arteries. The inhibitory effects of rotenone were reversed by exogenous hydrogen peroxide. A PKCepsilon translocation peptide inhibitor or PKCepsilon gene deletion decreased hypoxic increase in [Ca(2+)](i) in PASMCs, whereas the conventional PKC inhibitor GO6976 had no effect. These data suggest that acute hypoxia may specifically increase mitochondrial ROS generation, which subsequently activates PKC, particularly PKCepsilon, contributing to hypoxia-induced increase in [Ca(2+)](i) and contraction in PASMCs.
Collapse
Affiliation(s)
- Rakesh Rathore
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208; and
| | - Yun-Min Zheng
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208; and
| | - Xiao-Qiang Li
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208; and
| | - Qing-Song Wang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208; and
| | - Qing-Hua Liu
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208; and
| | - Roman R. Ginnan
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208; and
| | - Harold A Singer
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208; and
| | - Ye-Shih Ho
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI 48201
| | - Yong-Xiao Wang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208; and
- *Corresponding author Fax: +1 518 2628101 E-mail address: (YX Wang)
| |
Collapse
|
38
|
Waypa GB, Guzy R, Mungai PT, Mack MM, Marks JD, Roe MW, Schumacker PT. Increases in mitochondrial reactive oxygen species trigger hypoxia-induced calcium responses in pulmonary artery smooth muscle cells. Circ Res 2006; 99:970-8. [PMID: 17008601 DOI: 10.1161/01.res.0000247068.75808.3f] [Citation(s) in RCA: 144] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mitochondria have been implicated as a potential site of O(2) sensing underlying hypoxic pulmonary vasoconstriction (HPV), but 2 disparate models have been proposed to explain their reaction to hypoxia. One model proposes that hypoxia-induced increases in mitochondrial reactive oxygen species (ROS) generation activate HPV through an oxidant-signaling pathway, whereas the other proposes that HPV is a result of decreased oxidant signaling. In an attempt to resolve this debate, we use a novel, ratiometric, redox-sensitive fluorescence resonance energy transfer (HSP-FRET) probe, in concert with measurements of reduced/oxidized glutathione (GSH/GSSG), to assess cytosolic redox responses in cultured pulmonary artery smooth muscle cells (PASMCs). Superfusion of PASMCs with hypoxic media increases the HSP-FRET ratio and decreases GSH/GSSG, indicating an increase in oxidant stress. The antioxidants pyrrolidinedithiocarbamate and N-acetyl-l-cysteine attenuated this response, as well as the hypoxia-induced increases in cytosolic calcium ([Ca(2+)](i)), assessed by the Ca(2+)-sensitive FRET sensor YC2.3. Adenoviral overexpression of glutathione peroxidase or cytosolic or mitochondrial catalase attenuated the hypoxia-induced increase in ROS signaling and [Ca(2+)](i). Adenoviral overexpression of cytosolic Cu, Zn-superoxide dismutase (SOD-I) had no effect on the hypoxia-induced increase in ROS signaling and [Ca(2+)](i), whereas mitochondrial matrix-targeted Mn-SOD (SOD-II) augmented [Ca(2+)](i). The mitochondrial inhibitor myxothiazol attenuated the hypoxia-induced changes in the ROS signaling and [Ca(2+)](i), whereas cyanide augmented the increase in [Ca(2+)](i). Finally, simultaneous measurement of ROS and Ca(2+) signaling in the same cell revealed that the initial increase in these 2 signals could not be distinguished temporally. These results demonstrate that hypoxia triggers increases in PASMC [Ca(2+)](i) by augmenting ROS signaling from the mitochondria.
Collapse
Affiliation(s)
- Gregory B Waypa
- Department of Pediatrics, Division of Neonatology, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Hoffman DL, Salter JD, Brookes PS. Response of mitochondrial reactive oxygen species generation to steady-state oxygen tension: implications for hypoxic cell signaling. Am J Physiol Heart Circ Physiol 2006; 292:H101-8. [PMID: 16963616 DOI: 10.1152/ajpheart.00699.2006] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mitochondria are proposed to play an important role in hypoxic cell signaling. One currently accepted signaling paradigm is that the mitochondrial generation of reactive oxygen species (ROS) increases in hypoxia. This is paradoxical, because oxygen is a substrate for ROS generation. Although the response of isolated mitochondrial ROS generation to [O(2)] has been examined previously, such investigations did not apply rigorous control over [O(2)] within the hypoxic signaling range. With the use of open-flow respirometry and fluorimetry, the current study determined the response of isolated rat liver mitochondrial ROS generation to defined steady-state [O(2)] as low as 0.1 microM. In mitochondria respiring under state 4 (quiescent) or state 3 (ATP turnover) conditions, decreased ROS generation was always observed at low [O(2)]. It is concluded that the biochemical mechanism to facilitate increased ROS generation in response to hypoxia in cells is not intrinsic to the mitochondrial respiratory chain alone but may involve other factors. The implications for hypoxic cell signaling are discussed.
Collapse
Affiliation(s)
- David L Hoffman
- Box 604 Anesthesiology, Univ. of Rochester Medical Center, 601 Elmwood Ave., Rochester, NY 14642, USA
| | | | | |
Collapse
|
40
|
Paddenberg R, König P, Faulhammer P, Goldenberg A, Pfeil U, Kummer W. Hypoxic vasoconstriction of partial muscular intra-acinar pulmonary arteries in murine precision cut lung slices. Respir Res 2006; 7:93. [PMID: 16808843 PMCID: PMC1524949 DOI: 10.1186/1465-9921-7-93] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Accepted: 06/29/2006] [Indexed: 11/10/2022] Open
Abstract
Background Acute alveolar hypoxia causes pulmonary vasoconstriction (HPV) which serves to match lung perfusion to ventilation. The underlying mechanisms are not fully resolved yet. The major vascular segment contributing to HPV, the intra-acinar artery, is mostly located in that part of the lung that cannot be selectively reached by the presently available techniques, e.g. hemodynamic studies of isolated perfused lungs, recordings from dissected proximal arterial segments or analysis of subpleural vessels. The aim of the present study was to establish a model which allows the investigation of HPV and its underlying mechanisms in small intra-acinar arteries. Methods Intra-acinar arteries of the mouse lung were studied in 200 μm thick precision-cut lung slices (PCLS). The organisation of the muscle coat of these vessels was characterized by α-smooth muscle actin immunohistochemistry. Basic features of intra-acinar HPV were characterized, and then the impact of reactive oxygen species (ROS) scavengers, inhibitors of the respiratory chain and Krebs cycle metabolites was analysed. Results Intra-acinar arteries are equipped with a discontinuous spiral of α-smooth muscle actin-immunoreactive cells. They exhibit a monophasic HPV (medium gassed with 1% O2) that started to fade after 40 min and was lost after 80 min. This HPV, but not vasoconstriction induced by the thromboxane analogue U46619, was effectively blocked by nitro blue tetrazolium and diphenyleniodonium, indicating the involvement of ROS and flavoproteins. Inhibition of mitochondrial complexes II (3-nitropropionic acid, thenoyltrifluoroacetone) and III (antimycin A) specifically interfered with HPV, whereas blockade of complex IV (sodium azide) unspecifically inhibited both HPV and U46619-induced constriction. Succinate blocked HPV whereas fumarate had minor effects on vasoconstriction. Conclusion This study establishes the first model for investigation of basic characteristics of HPV directly in intra-acinar murine pulmonary vessels. The data are consistent with a critical involvement of ROS, flavoproteins, and of mitochondrial complexes II and III in intra-acinar HPV. In view of the lack of specificity of any of the classical inhibitors used in such types of experiments, validation awaits the use of appropriate knockout strains and siRNA interference, for which the present model represents a well-suited approach.
Collapse
Affiliation(s)
- Renate Paddenberg
- University of Giessen Lung Center, Institute for Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Peter König
- University of Giessen Lung Center, Institute for Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Petra Faulhammer
- University of Giessen Lung Center, Institute for Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Anna Goldenberg
- University of Giessen Lung Center, Institute for Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Uwe Pfeil
- University of Giessen Lung Center, Institute for Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Wolfgang Kummer
- University of Giessen Lung Center, Institute for Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
41
|
Ward JPT. Point: Hypoxic pulmonary vasoconstriction is mediated by increased production of reactive oxygen species. J Appl Physiol (1985) 2006; 101:993-5; discussion 999. [PMID: 16675614 DOI: 10.1152/japplphysiol.00480.2006] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Jeremy P T Ward
- Division of Asthma, Allergy and Lung Biology, King's College London School of Medicine, UK.
| |
Collapse
|
42
|
Sarafian TA, Habib N, Oldham M, Seeram N, Lee RP, Lin L, Tashkin DP, Roth MD. Inhaled marijuana smoke disrupts mitochondrial energetics in pulmonary epithelial cells in vivo. Am J Physiol Lung Cell Mol Physiol 2006; 290:L1202-9. [PMID: 16414979 DOI: 10.1152/ajplung.00371.2005] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Habitual marijuana smoking is associated with inflammation and atypia of airway epithelium accompanied by symptoms of chronic bronchitis. We hypothesized that Delta(9)-tetrahydrocannabinol (THC), the primary psychoactive component of marijuana, might contribute to these findings by impairing cellular energetics and mitochondrial function. To test this hypothesis, we examined particulate smoke extracts from marijuana cigarettes, tobacco cigarettes, and placebo marijuana (0% THC) cigarettes for their effects on the mitochondrial function of A549 cells in vitro. Only extracts prepared from marijuana cigarettes altered mitochondrial staining by the potentiometric probe JC-1. With the use of a cross-flow, nose-only inhalation system, rats were then exposed for 20 min to whole marijuana smoke and examined for its effects on airway epithelial cells. Inhalation of marijuana smoke produced lung tissue concentrations of THC that were 8-10 times higher than those measured in blood (75 +/- 38 ng/g wet wt tissue vs. 9.2 +/- 2.0 ng/ml), suggesting high local exposure. Intratracheal infusion of JC-1 immediately following marijuana smoke exposure revealed a diffuse decrease in lung cell JC-1 red fluorescence compared with tissue from unexposed or placebo smoke-exposed rats. Exposure to marijuana smoke in vivo also decreased JC-1 red fluorescence (54% decrease, P < 0.01) and ATP levels (75% decrease, P < 0.01) in single-cell preparations of tracheal epithelial cells. These results suggest that inhalation of marijuana smoke has deleterious effects on airway epithelial cell energetics that may contribute to the adverse pulmonary consequences of marijuana smoking.
Collapse
Affiliation(s)
- Theodore A Sarafian
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Weissmann N, Zeller S, Schäfer RU, Turowski C, Ay M, Quanz K, Ghofrani HA, Schermuly RT, Fink L, Seeger W, Grimminger F. Impact of mitochondria and NADPH oxidases on acute and sustained hypoxic pulmonary vasoconstriction. Am J Respir Cell Mol Biol 2005; 34:505-13. [PMID: 16357364 DOI: 10.1165/rcmb.2005-0337oc] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
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.
Collapse
Affiliation(s)
- Norbert Weissmann
- University Giessen Lung Centre (UGLC), Medical Clinic II/V, Justus-Liebig-University Giessen, Klinikstrasse 36, 35392 Giessen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Liu JQ, Zelko IN, Erbynn EM, Sham JSK, Folz RJ. Hypoxic pulmonary hypertension: role of superoxide and NADPH oxidase (gp91phox). Am J Physiol Lung Cell Mol Physiol 2005; 290:L2-10. [PMID: 16085672 DOI: 10.1152/ajplung.00135.2005] [Citation(s) in RCA: 225] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic exposure to low-O2 tension induces pulmonary arterial hypertension (PAH), which is characterized by vascular remodeling and enhanced vasoreactivity. Recent evidence suggests that reactive oxygen species (ROS) may be involved in both processes. In this study, we critically examine the role superoxide and NADPH oxidase plays in the development of chronic hypoxic PAH. Chronic hypoxia (CH; 10% O2 for 3 wk) caused a significant increase in superoxide production in intrapulmonary arteries (IPA) of wild-type (WT) mice as measured by lucigenin-enhanced chemiluminescence. The CH-induced increase in the generation of ROS was obliterated in NADPH oxidase (gp91phox) knockout (KO) mice, suggesting that NADPH oxidase was the major source of ROS. Importantly, pathological changes associated with CH-induced PAH (mean right ventricular pressure, medial wall thickening of small pulmonary arteries, and right heart hypertrophy) were completely abolished in NADPH oxidase (gp91phox) KO mice. CH potentiated vasoconstrictor responses of isolated IPAs to both 5-hydroxytryptamine (5-HT) and the thromboxane mimetic U-46619. Administration of CuZn superoxide dismutase to isolated IPA significantly reduced CH-enhanced superoxide levels and reduced the CH-enhanced vasoconstriction to 5-HT and U-46619. Additionally, CH-enhanced superoxide production and vasoconstrictor activity seen in WT IPAs were markedly reduced in IPAs isolated from NADPH oxidase (gp91phox) KO mice. These results demonstrate a pivotal role for gp91phox-dependent superoxide production in the pathogenesis of CH-induced PAH.
Collapse
Affiliation(s)
- John Q Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, MSRB 341, Durham, NC 27710, USA.
| | | | | | | | | |
Collapse
|
45
|
Abstract
Recently, the mitochondria have become the focus of attention as the site of O(2) sensing underlying hypoxic pulmonary vasoconstriction (HPV). However, two disparate models have emerged to explain how mitochondria react to a decrease in Po(2). One model proposes that a drop in Po(2) decreases the rate of mitochondrial reactive oxygen species (ROS) generation, resulting in a decrease in oxidant stress and an accumulation of reducing equivalents. The resulting shift of the cytosol to a reduced state causes the inhibition of voltage-dependent potassium channels, membrane depolarization, and the influx of calcium through voltage-gated (L-type) calcium channels. A second and opposing model suggests that hypoxia triggers a paradoxical increase in a mitochondrial-induced ROS signal. The resulting shift of the cytosol to an oxidized state triggers the release of intracellular calcium stores, recruitment of calcium channels in the plasma membrane, and activation of contraction. This article summarizes the potential involvement of a mitochondria-induced ROS signal in these two very different models.
Collapse
Affiliation(s)
- Gregory B Waypa
- Dept. of Medicine MC6026, The University of Chicago, 5841 South Maryland Ave., Chicago, IL 60637, USA
| | | |
Collapse
|