Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Antioxidants (Basel) ; 12(11)2023 Nov 02.
Article in English | MEDLINE | ID: mdl-38001814

ABSTRACT

BACKGROUND AND AIMS: we previously reported in studies on organoid-cultured bovine pulmonary arteries that pulmonary hypertension (PH) conditions of exposure to hypoxia or endothelin-1 caused a loss of a cartilage oligomeric matrix protein (COMP) stabilization of bone morphogenetic protein receptor-2 (BMPR2) function, a known key process contributing to pulmonary hypertension development. Based on subsequent findings, these conditions were associated with an extracellular superoxide-mediated increase in matrix metalloproteinase 9 (MMP-9) expression. We investigated if this contributed to PH development using mice deficient in MMP9. RESULTS: wild-type (WT) mice exposed to Sugen/Hypoxia (SuHx) to induce PH had increased levels of MMP9 in their lungs. Hemodynamic measures from MMP9 knockout mice (MMP9 KO) indicated they had attenuated PH parameters compared to WT mice based on an ECHO assessment of pulmonary artery pressure, right ventricular systolic pressure, and Fulton index hypertrophy measurements. In vitro vascular reactivity studies showed impaired endothelium-dependent and endothelium-independent NO-associated vasodilatory responses in the pulmonary arteries of SuHx mice and decreased lung levels of COMP and BMPR2 expression. These changes were attenuated in MMP9 KO mice potentially through preserving COMP-dependent stabilization of BMPR2. INNOVATION: this study supports a new function of superoxide in increasing MMP9 and the associated impairment of BMPR2 in promoting PH development which could be a target for future therapies. CONCLUSION: superoxide, through promoting increases in MMP9, mediates BMPR2 depletion and its consequent control of vascular function in response to PH mediators and the SuHx mouse model of PH.

2.
Am J Physiol Lung Cell Mol Physiol ; 323(4): L400-L409, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-35943724

ABSTRACT

This study examines if heme biosynthesis-associated iron metabolism is regulated in pulmonary arteries by endothelin-1 (ET1) potentially through modulating cartilage oligomeric matrix protein (COMP) availability. Our studies in organoid-cultured endothelium-rubbed bovine pulmonary arteries (BPAs) observed COMP depletion by siRNA or hypoxia increases NOX2 and superoxide and depletes mitochondrial SOD2. ET1 also increases superoxide in a manner that potentially impairs mitochondrial heme biosynthesis. In this study, organoid culture of BPA with ET1 (10 nM) increases superoxide in the mitochondrial matrix and extramitochondrial regions associated with COMP depletion, and COMP (0.5 µM) inhibited these superoxide increases. As mitochondrial matrix superoxide could impair heme biosynthesis from protoporphyrin IX (PpIX) by decreasing Fe2+ availability and/or ferrochelatase (FECH), we studied ET1, COMP, and COMP siRNA effects on the expression of FECH, transferrin receptor-1 (TfR1, an indicator of iron availability) and soluble guanylate cyclase (sGC, a key heme-dependent protein), and on measurements of PpIX (HPLC) and heme content. ET1 decreased FECH, heme, and sGC, and increased TfR1 and iron. COMP reversed these effects of ET1, and COMP decreased PpIX and increased heme in the absence of ET1. COMP siRNA increased PpIX detection and TfR1 expression and decreased the expression of FECH and sGC. Nitric oxide (spermine NONOate) relaxation of BPA was inhibited by ET1, and this was attenuated by COMP during exposure to ET1. Thus, COMP depletion by ET1 or siRNA modulates pulmonary artery iron metabolism, which results in loss of heme biosynthesis and heme-dependent cGMP mechanisms.


Subject(s)
Pulmonary Artery , Superoxides , Animals , Cartilage Oligomeric Matrix Protein/genetics , Cattle , Endothelin-1/metabolism , Ferrochelatase/metabolism , Ferrochelatase/pharmacology , Heme/metabolism , Iron/metabolism , Nitric Oxide/metabolism , Pulmonary Artery/metabolism , RNA, Small Interfering/metabolism , Receptors, Transferrin/metabolism , Soluble Guanylyl Cyclase/metabolism , Superoxides/metabolism
3.
Am J Physiol Lung Cell Mol Physiol ; 317(5): L569-L577, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31389735

ABSTRACT

Changes in reactive oxygen species and extracellular matrix seem to participate in pulmonary hypertension development. Because we recently reported evidence for chronic hypoxia decreasing expression of cartilage oligomeric matrix protein (COMP) and evidence for this controlling loss of pulmonary arterial smooth muscle bone morphogenetic protein receptor-2 (BMPR2) and contractile phenotype proteins, we examined if changes in superoxide metabolism could be an important factor in a bovine pulmonary artery (BPA), organoid cultured under hypoxia for 48 h model. Hypoxia (3% O2) caused a depletion of COMP in BPA, but not in bovine coronary arteries. Knockdown of COMP by small-interfering RNA (siRNA) increased BPA levels of mitochondrial and extra-mitochondrial superoxide detected by MitoSOX and dihydroethidium (DHE) HPLC products. COMP siRNA-treated BPA showed reduced levels of SOD2 and SOD3 and increased levels of NADPH oxidases NOX2 and NOX4. Hypoxia increased BPA levels of MitoSOX-detected superoxide and caused changes in NOX2 and SOD2 expression similar to COMP siRNA, and exogenous COMP (0.5 µM) prevented the effects of hypoxia. In the presence of COMP, BMPR2 siRNA-treated BPA showed increases in superoxide detected by MitoSOX and depletion of SOD2. Superoxide scavengers (0.5 µM TEMPO or mitoTEMPO) maintained the expression of contractile phenotype proteins calponin and SM22α decreased by 48 h hypoxia (1% O2). Adenoviral delivery of BMPR2 to rat pulmonary artery smooth muscle cells prevented the depletion of calponin and SM22α by COMP siRNA. Thus, COMP regulation of BMPR2 appears to have an important role in controlling hypoxia-elicited changes in BPA superoxide and its potential regulation of contractile phenotype proteins.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/genetics , Cartilage Oligomeric Matrix Protein/genetics , Hypoxia/genetics , Myocytes, Smooth Muscle/drug effects , Oxygen/pharmacology , Superoxides/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type II/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cartilage Oligomeric Matrix Protein/antagonists & inhibitors , Cartilage Oligomeric Matrix Protein/metabolism , Cattle , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Gene Expression Regulation , Heart/drug effects , Hypoxia/metabolism , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Primary Cell Culture , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tissue Culture Techniques , Calponins
4.
Am J Physiol Heart Circ Physiol ; 310(11): H1439-47, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27037373

ABSTRACT

Oxidation of the soluble guanylate cyclase (sGC) heme promotes loss of regulation by nitric oxide (NO) and depletion of sGC. We hypothesized that angiotensin II (ANG II) stimulation of mitochondrial superoxide by its type 1 receptor could function as a potential inhibitor of heme biosynthesis by ferrochelatase, and this could decrease vascular responsiveness to NO by depleting sGC. These processes were investigated in a 24-h organoid culture model of bovine coronary arteries (BCA) with 0.1 µM ANG II. Treatment of BCA with ANG II increased mitochondrial superoxide, depleted mitochondrial superoxide dismutase (SOD2), ferrochelatase, and cytochrome oxidase subunit 4, and sGC, associated with impairment of relaxation to NO. These processes were attenuated by organoid culture with 8-bromo-cGMP and/or δ-aminolevulinic acid (a stimulator of sGC by protoporphyrin IX generation and heme biosynthesis). Organoid culture with Mito-TEMPOL, a scavenger of mitochondrial matrix superoxide, also attenuated ANG II-elicited ferrochelatase depletion and loss of relaxation to NO, whereas organoid culture with Tempol, an extramitochondrial scavenger of superoxide, attenuated the loss of relaxation to NO by ANG II, but not ferrochelatase depletion, suggesting cytosolic superoxide could be an initiating factor in the loss of sGC regulation by NO. The depletion of cytochrome oxidase subunit 4 and sGC (but not catalase) suggests that sGC expression may be very sensitive to depletion of heme caused by ANG II disrupting ferrochelatase activity by increasing mitochondrial superoxide. In addition, cGMP-dependent activation of protein kinase G appears to attenuate these ANG II-stimulated processes through both preventing SOD2 depletion and increases in mitochondrial and extramitochondrial superoxide.


Subject(s)
Angiotensin II/pharmacology , Coronary Vessels/drug effects , Ferrochelatase/metabolism , Heme/metabolism , Mitochondria/drug effects , Soluble Guanylyl Cyclase/metabolism , Superoxides/metabolism , Animals , Cattle , Coronary Vessels/enzymology , Cyclic GMP-Dependent Protein Kinases/metabolism , Down-Regulation , Electron Transport Complex IV/metabolism , Enzyme Activators/pharmacology , Free Radical Scavengers/pharmacology , Mitochondria/enzymology , Nitric Oxide/metabolism , Nitric Oxide Donors/metabolism , Nitric Oxide Donors/pharmacology , Organoids , Superoxide Dismutase/metabolism , Tissue Culture Techniques , Vasodilation/drug effects , Vasodilator Agents/pharmacology
5.
Am J Physiol Lung Cell Mol Physiol ; 308(7): L719-28, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25659899

ABSTRACT

This study examines how heme biosynthesis modulation with δ-aminolevulinic acid (ALA) potentially functions to prevent 21-day hypoxia (10% oxygen)-induced pulmonary hypertension in mice and the effects of 24-h organoid culture with bovine pulmonary arteries (BPA) with the hypoxia and pulmonary hypertension mediator endothelin-1 (ET-1), with a focus on changes in superoxide and regulation of micro-RNA 204 (miR204) expression by src kinase phosphorylation of signal transducer and activator of transcription-3 (STAT3). The treatment of mice with ALA attenuated pulmonary hypertension (assessed through echo Doppler flow of the pulmonary valve, and direct measurements of right ventricular systolic pressure and right ventricular hypertrophy), increases in pulmonary arterial superoxide (detected by lucigenin), and decreases in lung miR204 and mitochondrial superoxide dismutase (SOD2) expression. ALA treatment of BPA attenuated ET-1-induced increases in mitochondrial superoxide (detected by MitoSox), STAT3 phosphorylation, and decreases in miR204 and SOD2 expression. Because ALA increases BPA protoporphyrin IX (a stimulator of guanylate cyclase) and cGMP-mediated protein kinase G (PKG) activity, the effects of the PKG activator 8-bromo-cGMP were examined and found to also attenuate the ET-1-induced increase in superoxide. ET-1 increased superoxide production and the detection of protoporphyrin IX fluorescence, suggesting oxidant conditions might impair heme biosynthesis by ferrochelatase. However, chronic hypoxia actually increased ferrochelatase activity in mouse pulmonary arteries. Thus, a reversal of factors increasing mitochondrial superoxide and oxidant effects that potentially influence remodeling signaling related to miR204 expression and perhaps iron availability needed for the biosynthesis of heme by the ferrochelatase reaction could be factors in the beneficial actions of ALA in pulmonary hypertension.


Subject(s)
Aminolevulinic Acid/pharmacology , Heme/biosynthesis , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/metabolism , Hypoxia/metabolism , Aminolevulinic Acid/therapeutic use , Animals , Endothelin-1/physiology , Ferrochelatase/metabolism , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypertrophy, Right Ventricular/etiology , Hypoxia/complications , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/enzymology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Superoxide Dismutase/metabolism , Superoxides/metabolism
6.
Am J Physiol Heart Circ Physiol ; 298(6): H1879-88, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20304813

ABSTRACT

Hypoxia appears to promote contraction [hypoxic pulmonary vasoconstriction (HPV)] of bovine pulmonary arteries (BPA) through removal of a peroxide-mediated relaxation. This study examines the roles of BPA Nox oxidases and mitochondria in the HPV response. Inhibitors of Nox2 (0.1 mM apocynin and 50 muM gp91-dstat) and mitochondrial electron transport (10 muM antimycin and rotenone) decreased superoxide generation in BPA without affecting contraction to 25 mM KCl or the HPV response. Transfection of BPA with small inhibitory RNA (siRNA) for Nox2 and Nox4 decreased Nox2 and Nox4 protein expression, respectively, associated with an attenuation of superoxide detection, without affecting 25 mM KCl contraction. However, Nox4 siRNA, but not Nox2, attenuated HPV in BPA. A Nox4 inhibitor plumbagin (10 muM) increased basal force, decreased superoxide detection and peroxide release, and caused BPA to relax under hypoxia. Although acute removal of peroxide with 0.1 mM ebselen increased 25 mM KCl contraction and decreased hypoxic contraction, prolonged treatment with ebselen only decreased hypoxic contraction without affecting 25 mM KCl contraction, suggesting basal peroxide levels also maintain a contractile mechanism not removed by acute hypoxia. Organ culture of BPA with transforming growth factor (TGF)-beta1 (4 nM) increased Nox4 expression, superoxide, peroxide, and the HPV response. Thus Nox2 and mitochondria are sources for superoxide generation in BPA, which do not appear to influence the HPV response. However, peroxide derived from superoxide generated by Nox4 appears to maintain a basal relaxation in BPA under normoxic conditions, which is removed under hypoxia leading to HPV. Peroxide generated by Nox4 may also function to maintain a contractile mechanism, which is not reversed by acute hypoxia.


Subject(s)
Hypoxia/physiopathology , NADPH Oxidases/physiology , Pulmonary Artery/physiopathology , Vasoconstriction/physiology , Animals , Antioxidants/pharmacology , Azoles/pharmacology , Cattle , Isoindoles , Mitochondria, Muscle/physiology , Models, Animal , Muscle, Smooth, Vascular/physiopathology , Organoselenium Compounds/pharmacology , Potassium Chloride/pharmacology , Reactive Oxygen Species/metabolism , Vasoconstriction/drug effects
7.
Am J Physiol Heart Circ Physiol ; 297(4): H1453-61, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19666846

ABSTRACT

Endothelium-denuded bovine pulmonary arteries (BPA) contract to hypoxia through a mechanism potentially involving removing a superoxide-derived hydrogen peroxide-mediated relaxation. BPA organ cultured for 24 h with 0.1 mM cobalt chloride (CoCl(2)) to increase the expression and activity of heme oxygenase-1 (HO-1) is accompanied by a decrease in 5 microM lucigenin-detectable superoxide and an increase in horseradish peroxidase-luminol detectable peroxide levels. Force development to KCl in BPA was not affected by increases in HO-1, but the hypoxic pulmonary vasoconstriction (HPV) response was decreased. Organ culture with a HO-1 inhibitor (10 microM chromium mesoporphyrin) reversed the effects of HO-1 on HPV and peroxide. Treatment of HO-1-induced BPA with extracellular catalase resulted in reversal of the attenuation of HPV without affecting the force development to KCl. Increasing intracellular peroxide scavenging with 0.1 mM ebselen increased force development to KCl and partially reversed the decrease in HPV seen on induction of HO-1. HO-1 induction increases extracellular (ec) superoxide dismutase (SOD) expression without changing Cu,Zn-SOD and Mn-SOD levels. HO-1-induced BPA rings treated with the copper chelator 10 mM diethyldithiocarbamate to inactivate ecSOD and Cu,Zn-SOD showed increased superoxide and decreased peroxide to levels equal to non-HO-1-induced rings, whereas the addition of SOD to freshly isolated BPA rings attenuated HPV similar to HO-1 induction with CoCl(2). Therefore, HO-1 induction in BPA increases ecSOD expression associated with enhanced generation of peroxide in amounts that may not be adequately removed during hypoxia, leading to an attenuation of HPV.


Subject(s)
Heme Oxygenase-1/metabolism , Pulmonary Artery/enzymology , Superoxide Dismutase/metabolism , Vasoconstriction , Animals , Azoles/pharmacology , Catalase/metabolism , Cattle , Cell Hypoxia , Chelating Agents/pharmacology , Cobalt/pharmacology , Copper/metabolism , Ditiocarb/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Heme Oxygenase-1/antagonists & inhibitors , Hydrogen Peroxide/metabolism , Isoindoles , Mesoporphyrins/pharmacology , Organ Culture Techniques , Organoselenium Compounds/pharmacology , Potassium Chloride/pharmacology , Pulmonary Artery/drug effects , Superoxides/metabolism , Up-Regulation , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
8.
Biochim Biophys Acta ; 1773(2): 185-91, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17052767

ABSTRACT

Exposure of platelets to toxins (calyculin A or okadaic acid) that inhibit protein serine/threonine phosphatases types 1 and 2A, at concentrations that block aggregatory and secretory responses, results in the phosphorylation of several platelet proteins including integrin beta(3). Since protein phosphorylation represents a balance between kinase and phosphatase activities, this increase in phosphorylation reflects either the removal of phosphatases that oppose constitutively active kinases known to reside in the platelet (e.g., casein kinase 2) or the activation of endogenous kinases. In this study, we demonstrate that the addition of calyculin A promotes the activation of several endogenous platelet protein kinases, including p42/44(mapk), p38(mapk), Akt/PKB, and LKB1. Using a pharmacologic approach, we assessed whether inhibition of these and other enzymes block phosphorylation of beta(3). Inhibitors of p38(mapk), casein kinase, AMP kinase, protein kinase C, and calcium-calmodulin-dependent kinases did not block phosphorylation of beta(3) on thr(753). In contrast, 5'-iodotubercidin, at 50 muM, blocks beta(3) phosphorylation without affecting the efficacy of calyculin A to inhibit platelet aggregation and spreading. These data dissociate threonine phosphorylation of beta(3) molecules and inhibition of platelet responses by protein phosphatase inhibitors.


Subject(s)
Blood Platelets/drug effects , Integrin beta3/metabolism , Oxazoles/pharmacology , Phosphothreonine/metabolism , Tubercidin/analogs & derivatives , 3-Phosphoinositide-Dependent Protein Kinases , AMP-Activated Protein Kinase Kinases , Amino Acid Motifs , Amino Acid Sequence , Blood Platelets/enzymology , Blood Platelets/metabolism , Conserved Sequence , Enzyme Activation/drug effects , Humans , Integrin beta3/chemistry , Marine Toxins , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Substrate Specificity/drug effects , Thrombin/pharmacology , Tubercidin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...