Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L303-16, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27233995

ABSTRACT

We (66) have previously described an NSAID-insensitive intramitochondrial biosynthetic pathway involving oxidation of the polyunsaturated mitochondrial phospholipid, cardiolipin (CL), followed by hydrolysis [by calcium-independent mitochondrial calcium-independent phospholipase A2-γ (iPLA2γ)] of oxidized CL (CLox), leading to the formation of lysoCL and oxygenated octadecadienoic metabolites. We now describe a model system utilizing oxidative lipidomics/mass spectrometry and bioassays on cultured bovine pulmonary artery endothelial cells (BPAECs) to assess the impact of CLox that we show, in vivo, can be released to the extracellular space and may be hydrolyzed by lipoprotein-associated PLA2 (Lp-PLA2). Chemically oxidized liposomes containing bovine heart CL produced multiple oxygenated species. Addition of Lp-PLA2 hydrolyzed CLox and produced (oxygenated) monolysoCL and dilysoCL and oxidized octadecadienoic metabolites including 9- and 13-hydroxyoctadecadienoic (HODE) acids. CLox caused BPAEC necrosis that was exacerbated by Lp-PLA2 Lower doses of nonlethal CLox increased permeability of BPAEC monolayers. This effect was exacerbated by Lp-PLA2 and partially mimicked by authentic monolysoCL or 9- or 13-HODE. Control mice plasma contained virtually no detectable CLox; in contrast, 4 h after Pseudomonas aeruginosa (P. aeruginosa) infection, 34 ± 8 mol% (n = 6; P < 0.02) of circulating CL was oxidized. In addition, molar percentage of monolysoCL increased twofold after P. aeruginosa in a subgroup analyzed for these changes. Collectively, these studies suggest an important role for 1) oxidation of CL in proinflammatory environments and 2) possible hydrolysis of CLox in extracellular spaces producing lysoCL and oxidized octadecadienoic acid metabolites that may lead to impairment of pulmonary endothelial barrier function and necrosis.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/physiology , Cardiolipins/biosynthesis , Endothelial Cells/physiology , Animals , Cardiolipins/blood , Cattle , Cells, Cultured , Electric Impedance , Hydrolysis , Mice, Inbred C57BL , Oxidation-Reduction , Pseudomonas Infections/blood , Signal Transduction
2.
PLoS One ; 8(11): e81903, 2013.
Article in English | MEDLINE | ID: mdl-24312378

ABSTRACT

Although a critical role for caveolae-mediated albumin transcytosis in pulmonary endothelium is well established, considerably less is known about caveolae-independent pathways. In this current study, we confirmed that cultured rat pulmonary microvascular (RPMEC) and pulmonary artery (RPAEC) endothelium endocytosed Alexa488-labeled albumin in a saturable, temperature-sensitive mode and internalization resulted in co-localization by fluorescence microscopy with cholera B toxin and caveolin-1. Although siRNA to caveolin-1 (cav-1) in RPAEC significantly inhibited albumin uptake, a remnant portion of albumin uptake was cav-1-independent, suggesting alternative pathways for albumin uptake. Thus, we isolated and cultured mouse lung endothelial cells (MLEC) from wild type and cav-1(-/-) mice and noted that ~ 65% of albumin uptake, as determined by confocal imaging or live cell total internal reflectance fluorescence microscopy (TIRF), persisted in total absence of cav-1. Uptake of colloidal gold labeled albumin was evaluated by electron microscopy and demonstrated that albumin uptake in MLEC from cav-1(-/-) mice was through caveolae-independent pathway(s) including clathrin-coated pits that resulted in endosomal accumulation of albumin. Finally, we noted that albumin uptake in RPMEC was in part sensitive to pharmacological agents (amiloride [sodium transport inhibitor], Gö6976 [protein kinase C inhibitor], and cytochalasin D [inhibitor of actin polymerization]) consistent with a macropinocytosis-like process. The amiloride sensitivity accounting for macropinocytosis also exists in albumin uptake by both wild type and cav-1(-/-) MLEC. We conclude from these studies that in addition to the well described caveolar-dependent pulmonary endothelial cell endocytosis of albumin, a portion of overall uptake in pulmonary endothelial cells is cav-1 insensitive and appears to involve clathrin-mediated endocytosis and macropinocytosis-like process.


Subject(s)
Albumins/metabolism , Caveolae/physiology , Endocytosis , Endothelium, Vascular/metabolism , Lung/blood supply , Animals , Base Sequence , Caveolin 1/genetics , Caveolin 1/metabolism , Cells, Cultured , DNA Primers , Endothelium, Vascular/cytology , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Pinocytosis , RNA, Small Interfering/genetics , Rats
3.
Am J Physiol Lung Cell Mol Physiol ; 304(5): L350-60, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23275622

ABSTRACT

Hypozincemia, with hepatic zinc accumulation at the expense of other organs, occurs in infection, inflammation, and aseptic lung injury. Mechanisms underlying zinc partitioning or its impact on extrahepatic organs are unclear. Here we show that the major zinc-binding protein, metallothionein (MT), is critical for zinc transmigration from lung to liver during hyperoxia and preservation of intrapulmonary zinc during hyperoxia is associated with an injury-resistant phenotype in MT-null mice. Particularly, lung-to-liver zinc ratios decreased in wild-type (WT) and increased significantly in MT-null mice breathing 95% oxygen for 72 h. Compared with female adult WT mice, MT-null mice were significantly protected against hyperoxic lung injury indicated by reduced inflammation and interstitial edema, fewer necrotic changes to distal airway epithelium, and sustained lung function at 72 h hyperoxia. Lungs of MT-null mice showed decreased levels of immunoreactive LC3, an autophagy marker, compared with WT mice. Analysis of superoxide dismutase (SOD) activity in the lungs revealed similar levels of manganese-SOD activity between strains under normoxia and hyperoxia. Lung extracellular SOD activity decreased significantly in both strains at 72 h of hyperoxia, although there was no difference between strains. Copper-zinc-SOD activity was ~4× higher under normoxic conditions in MT-null compared with WT mice but was not affected in either group by hyperoxia. Collectively the data suggest that genetic deletion of MT-I/II in mice is associated with compensatory increase in copper-zinc-SOD activity, prevention of hyperoxia-induced zinc transmigration from lung to liver, and hyperoxia-resistant phenotype strongly associated with differences in zinc homeostasis during hyperoxic acute lung injury.


Subject(s)
Acute Lung Injury/metabolism , Liver/metabolism , Lung/metabolism , Metallothionein/metabolism , Superoxide Dismutase/metabolism , Zinc/metabolism , Animals , Female , Hyperoxia , Inflammation/immunology , Metallothionein/genetics , Mice , Mice, Knockout , Microtubule-Associated Proteins/analysis , Respiratory Mucosa/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 300(4): L526-33, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21239532

ABSTRACT

S-nitrosoalbumin (SNO-Alb) has been shown to be an efficacious cytoprotective molecule in acute lung injury, as well as ischemia-reperfusion injury in heart and skeletal muscle. Nonetheless, limited information is available on the cellular mechanism of such protection. Accordingly, we investigated the protective effects of SNO-Alb [ and its denitrosated congener, reduced albumin (SH-Alb) ] on tert-butyl hydroperoxide (tBH)-mediated cytotoxicity in cultured rat pulmonary microvascular endothelial cells (RPMEC), as well as hydrogen sulfide (H(2)S)-mediated cytotoxicity in rat pulmonary artery smooth muscle cells (RPASMC). We noted that tBH caused a concentration-dependent necrosis in RPMEC, and pretreatment of RPMEC with SNO-Alb dose-dependently decreased the sensitivity of these cells to tBH. A component of SNO-Alb cytoprotection was sensitive to N(G)-nitro-L-arginine methyl ester and was associated with activation of endothelial nitric oxide synthase (eNOS), phenomena that could be reproduced with pretreatment with SH-Alb. Exogenous H(2)S caused concentration-dependent apoptosis in RPASMC due to activation of ERK1/2 and p38, as well as downregulation of Bcl-2. Pretreatment with SNO-Alb reduced H(2)S-mediated apoptosis in a concentration-dependent manner that was associated with SNO-Alb-mediated inhibition of activation of ERK1/2 and p38. Pretreatment with SNO-Alb reduced toxicity of 1 mM sodium hydrosulfide in an N(G)-nitro-L-arginine methyl ester-sensitive fashion in RPASMC that expressed gp60 and neuronal NOS and was capable of transporting fluorescently labeled SH-Alb. Therefore, SNO-Alb is cytoprotective against models of oxidant-induced necrosis (tBH) and inhibitors of cellular respiration and apoptosis (H(2)S) in both pulmonary endothelium and smooth muscle, respectively, and a component of such protection can be attributed to a SH-Alb-mediated activation of constitutive NOS.


Subject(s)
Cytoprotection/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Lung/blood supply , Nitroso Compounds/pharmacology , Serum Albumin, Bovine/pharmacology , Animals , Apoptosis/drug effects , Arteries/cytology , Cells, Cultured , Endocytosis/drug effects , Endothelial Cells/enzymology , Humans , Hydrogen Sulfide/pharmacology , Microvessels/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Necrosis/pathology , Nitric Oxide Synthase Type III/metabolism , Nitrosation/drug effects , Oxidation-Reduction/drug effects , Rats , tert-Butylhydroperoxide/pharmacology
5.
Am J Physiol Lung Cell Mol Physiol ; 300(4): L624-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21239534

ABSTRACT

A role in signal transduction for a vanishingly small labile pool of intracellular zinc ([Zn](i)) has been inferred by the sensitivity of various physiological pathways to zinc chelators such as N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN) and/or associations with changes in nonprotein-bound zinc-sensitive fluorophores. Although we (44) reported that LPS-induced apoptosis in cultured sheep pulmonary artery endothelial cells (SPAEC) was exacerbated by TPEN, 1) we did not detect acute (30 min) changes in [Zn](i), and 2) it is unclear from other reports whether LPS increases or decreases [Zn](i) and whether elevations or decreases in [Zn](i) are associated with cell death and/or apoptosis. In the present study, we used both chemical (FluoZin-3 via live cell epifluorescence microscopy and fluorescence-activated cell sorting) and genetic (luciferase activity of a chimeric reporter encoding zinc-sensitive metal-response element and changes in steady-state mRNA of zinc importer, SLC39A14 or ZIP14) techniques to show that LPS caused a delayed time-dependent (2-4 h) decrease in [Zn](i) in SPAEC. A contributory role of decreases in [Zn](i) in LPS-induced apoptosis (as determined by caspase-3/7 activation, annexin-V binding, and cytochrome c release) in SPAECs was revealed by mimicking the effect of LPS with the zinc chelator, TPEN, and inhibiting LPS- (or TPEN)-induced apoptosis with exogenous zinc. Collectively, these are the first data demonstrating a signaling role for decrease in [Zn](i) in pulmonary endothelial cells and suggest that endogenous levels of labile zinc may affect sensitivity of pulmonary endothelium to the important and complex proapoptotic stimulus of LPS.


Subject(s)
Apoptosis/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Intracellular Space/metabolism , Lipopolysaccharides/pharmacology , Pulmonary Artery/cytology , Zinc/metabolism , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Flow Cytometry , Genes, Reporter , Intracellular Space/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sheep , Signal Transduction/drug effects , Spectrometry, Fluorescence , Up-Regulation/drug effects
6.
Ann N Y Acad Sci ; 1203: 73-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20716286

ABSTRACT

We have shown that zinc-thiolate moieties of the metal binding protein metallothionein (MT) are critical targets for nitric oxide (NO) with resultant increases in intracellular labile zinc. Such an NO-MT-Zn signaling pathway appears to participate in important cardiovascular functions associated with biosynthesis of NO including hypoxic vasoconstriction in the lung. Although downstream effector signaling molecules and critical contractile targets remain unclear, current investigations reveal a contributory role for zinc dependent protein kinases and cytoskeletal proteins in mediating hypoxic induced constriction of pulmonary endothelial cells.


Subject(s)
Endothelium/cytology , Endothelium/physiology , Homeostasis/physiology , Lung/cytology , Lung/physiology , Nitric Oxide/physiology , Signal Transduction/physiology , Zinc/physiology , Animals , Cell Communication/physiology , Cell Death/physiology , Endothelium/metabolism , Humans , Lung/chemistry , Lung/metabolism , Metallothionein/physiology , Nitric Oxide/chemistry , Zinc/chemistry
7.
J Chromatogr B Analyt Technol Biomed Life Sci ; 877(26): 2863-72, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19328050

ABSTRACT

Oxidation of two anionic phospholipids--cardiolipin (CL) in mitochondria and phosphatidylserine (PS) in extramitochondrial compartments--is important signaling event, particularly during the execution of programmed cell death and clearance of apoptotic cells. Quantitative analysis of CL and PS oxidation products is central to understanding their molecular mechanisms of action. We combined the identification of diverse phospholipid molecular species by ESI-MS with quantitative assessments of lipid hydroperoxides using a fluorescence HPLC-based protocol. We characterized CL and PS oxidation products formed in a model system (cyt c/H(2)O(2)), in apoptotic cells (neurons, pulmonary artery endothelial cells) and mouse lung under inflammatory/oxidative stress conditions (hyperoxia, inhalation of single walled carbon nanotubes). Our results demonstrate the usefulness of this approach for quantitative assessments, identification of individual molecular species and structural characterization of anionic phospholipids that are involved in oxidative modification in cells and tissues.


Subject(s)
Apoptosis , Cardiolipins/chemistry , Cardiolipins/immunology , Phosphatidylserines/chemistry , Phosphatidylserines/immunology , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Cells, Cultured , Endothelial Cells/chemistry , Endothelial Cells/cytology , Endothelial Cells/immunology , Lipid Peroxides/chemistry , Lipid Peroxides/immunology , Male , Mice , Mice, Inbred C57BL , Models, Biological , Neurons/chemistry , Neurons/cytology , Neurons/immunology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Sheep
8.
Am J Respir Cell Mol Biol ; 41(1): 69-75, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19097988

ABSTRACT

We recently reported that induction of metallothionein (MT) was critical in limiting nickel (Ni)-induced lung injury in intact mice. Nonetheless, the mechanism by which Ni induces MT expression is unclear. We hypothesized that the ability of Ni to mobilize zinc (Zn) may contribute to such regulation and therefore, we examined the mechanism for Ni-induced MT2A expression in human airway epithelial (BEAS-2B) cells. Ni induced MT2A transcript levels and protein expression by 4 hours. Ni also increased the activity of a metal response element (MRE) promoter luciferase reporter construct, suggesting that Ni induces MRE binding of the metal transcription factor (MTF-1). Exposure to Ni resulted in the nuclear translocation of MTF-1, and Ni failed to induce MT in mouse embryonic fibroblasts lacking MTF-1. As Zn is the only metal known to directly bind MTF-1, we then showed that Ni increased a labile pool of intracellular Zn in cells as revealed by fluorescence-activated cell sorter using the Zn-sensitive fluorophore, FluoZin-3. Ni-induced increases in MT2A mRNA and MRE-luciferase activity were sensitive to the Zn chelator, TPEN, supporting an important role for Zn in mediating the effect of Ni. Although neither the source of labile Zn nor the mechanism by which Ni liberates labile Zn was apparent, it was noteworthy that Ni increased intracellular reactive oxygen species (ROS). Although both N-acetyl cysteine (NAC) and ascorbic acid (AA) decreased Ni-induced increases in ROS, only NAC prevented Ni-induced increases in MT2A mRNA, suggesting a special role for interactions of Ni, thiols, and Zn release.


Subject(s)
Bronchi/metabolism , Chlorides/toxicity , Epithelial Cells/drug effects , Metallothionein/metabolism , Nickel/toxicity , Zinc Compounds/toxicity , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Cell Separation/methods , Cells, Cultured , Chelating Agents/pharmacology , Chlorides/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Ethylamines/pharmacology , Flow Cytometry , Fluorescent Dyes , Humans , Metallothionein/genetics , Mice , Mice, Knockout , Polycyclic Compounds , Pyridines , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/drug effects , Transfection , Up-Regulation , Zinc Compounds/metabolism , Transcription Factor MTF-1
9.
Circ Res ; 102(12): 1575-83, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18483408

ABSTRACT

The metal binding protein metallothionein (MT) is a target for nitric oxide (NO), causing release of bound zinc that affects myogenic reflex in systemic resistance vessels. Here, we investigate a role for NO-induced zinc release in pulmonary vasoregulation. We show that acute hypoxia causes reversible constriction of intraacinar arteries (<50 microm/L) in isolated perfused mouse lung (IPL). We further demonstrate that isolated pulmonary (but not aortic) endothelial cells constrict in hypoxia. Hypoxia also causes NO-dependent increases in labile zinc in mouse lung endothelial cells and endothelium of IPL. The latter observation is dependent on MT because it is not apparent in IPL of MT(-/-) mice. Data from NO-sensitive fluorescence resonance energy transfer-based reporters support hypoxia-induced NO production in pulmonary endothelium. Furthermore, hypoxic constriction is blunted in IPL of MT(-/-) mice and in wild-type mice, or rats, treated with the zinc chelator N,N,N',N'-tetrakis(2-pyridylmethyl)-ethylenediamine (TPEN), suggesting a role for chelatable zinc in modulating HPV. Finally, the NO donor DETAnonoate causes further vasoconstriction in hypoxic IPL in which NO vasodilatory pathways are inhibited. Collectively, these data suggest that zinc thiolate signaling is a component of the effects of acute hypoxia-mediated NO biosynthesis and that this pathway may contribute to constriction in the pulmonary vasculature.


Subject(s)
Hypoxia/physiopathology , Nitric Oxide/physiology , Pulmonary Artery/drug effects , Vascular Resistance/drug effects , Zinc/physiology , Animals , Aorta/drug effects , Cell Size , Cells, Cultured , Chelating Agents/pharmacology , Endothelial Cells/drug effects , Ethylenediamines/pharmacology , In Vitro Techniques , Metallothionein/drug effects , Metallothionein/physiology , Mice , Mice, Transgenic , Microscopy, Fluorescence , Nitrosation , Organ Specificity , Oxygen/pharmacology , Rats , Rats, Sprague-Dawley , Sheep , Vasoconstriction/drug effects
10.
Free Radic Biol Med ; 37(6): 785-92, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15304254

ABSTRACT

Low-molecular-weight S-nitrosothiols are found in many tissues and affect a diverse array of signaling pathways via decomposition to *NO or exchange of their -NO function with thiol-containing proteins (transnitrosation). We used spectral laser scanning confocal imaging to visualize the effects of D- and L-stereoisomers of S-nitrosocysteine ethyl ester (SNCEE) on fluorescence resonance energy transfer (FRET)-based reporters that are targets for the following NO-related modifications: (a) S-nitrosation, via the cysteine-rich protein metallothionein (FRET-MT), and (b) nitrosyl-heme-Fe, via guanosine 3',5'-cyclic monophosphate (cygnet-2). Conformational changes consistent with S-nitrosation of FRET-MT were specific to l-SNCEE. In addition, they were reversed by dithiothreitol (DTT) but unaffected by exogenous oxyhemoglobin. In contrast, d- and l-SNCEE had comparable effects on cygnet-2, likely via activation of soluble guanylyl cyclase (sGC) by *NO as they were sensitive to the sGC inhibitor 1H-[1,2,4]-oxadiazolo[4,3-alpha] quinoxalin-1-one and exogenous oxyhemoglobin. These data demonstrate the utility of spectral laser scanning confocal imaging in revealing subtle aspects of NO signal transduction in live cells. Stereoselective transnitrosation of MT emphasizes the specificity of posttranslational modification as a component of NO signaling.


Subject(s)
Cysteine/analogs & derivatives , Microscopy, Fluorescence/methods , Nitric Oxide/metabolism , Spectrometry, Fluorescence/methods , Sulfhydryl Compounds/chemistry , Animals , Cells, Cultured , Cyclic GMP/metabolism , Cysteine/chemistry , Dithiothreitol/chemistry , Endothelium, Vascular/cytology , Enzyme Activation , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/pharmacology , Free Radicals , Genes, Reporter , Green Fluorescent Proteins/metabolism , Guanylate Cyclase , Heme/chemistry , Nitric Oxide/chemistry , Nitroso Compounds/chemistry , Peptides/chemistry , Protein Conformation , Proteins/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Recombinant Fusion Proteins/metabolism , Sheep , Signal Transduction , Soluble Guanylyl Cyclase
11.
Mol Cell Biochem ; 234-235(1-2): 211-7, 2002.
Article in English | MEDLINE | ID: mdl-12162436

ABSTRACT

We hypothesized that: (a) S-nitrosylation of metallothionein (MT) is a component of pulmonary endothelial cell nitric oxide (NO) signaling that is associated with an increase in labile zinc; and (b) NO mediated increases in labile zinc in turn reduce the sensitivity of pulmonary endothelium to LPS-induced apoptosis. We used microspectrofluorometric techniques to show that exposing mouse lung endothelial cells (MLEC) to the NO-donor, S-nitrosocysteine, resulted in a 45% increase in fluorescence of the Zn2+-specific fluorophore, Zinquin, that was rapidly reversed by exposure to the Zn2+ chelator, NNN'N'-tetrakis-(2-pyridylmethyl)ethylenediamine; TPEN). The absence of a NO-mediated increase in labile Zn2+ in MLEC from MT-I and -II knockout mice inferred a critical role for MT in the regulation of Zn2+ homeostasis by NO. Furthermore, we found that prior exposure of cultured endothelial cells from sheep pulmonary artery (SPAEC), to the NO-donor, S-nitroso-N-acetylpenicillamine (SNAP) reduced their sensitivity to lipopolysaccharide (LPS) induced apoptosis. The anti-apoptotic effects of NO were significantly inhibited by Zn2+ chelation with low doses of TPEN (10 microM). Collectively, these data suggest that S-nitrosylation of MT is associated with an increase in labile (TPEN chelatable) zinc and NO-mediated MT dependent zinc release is associated with reduced sensitivity to LPS-induced apoptosis in pulmonary endothelium.


Subject(s)
Apoptosis/drug effects , Endothelium/cytology , Endothelium/drug effects , Lipopolysaccharides/pharmacology , Nitric Oxide/pharmacology , Zinc/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Endothelium/metabolism , Homeostasis/drug effects , Lung , Metallothionein/metabolism , Mice , Mice, Knockout , Microscopy, Fluorescence , Nitric Oxide/metabolism , Pulmonary Artery , Sheep , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...