Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Adv Exp Med Biol ; 1304: 333-373, 2021.
Article in English | MEDLINE | ID: mdl-34019276

ABSTRACT

Protein folding overload and oxidative stress disrupt endoplasmic reticulum (ER) homeostasis, generating reactive oxygen species (ROS) and activating the unfolded protein response (UPR). The altered ER redox state induces further ROS production through UPR signaling that balances the cell fates of survival and apoptosis, contributing to pulmonary microvascular inflammation and dysfunction and driving the development of pulmonary hypertension (PH). UPR-induced ROS production through ER calcium release along with NADPH oxidase activity results in endothelial injury and smooth muscle cell (SMC) proliferation. ROS and calcium signaling also promote endothelial nitric oxide (NO) synthase (eNOS) uncoupling, decreasing NO production and increasing vascular resistance through persistent vasoconstriction and SMC proliferation. C/EBP-homologous protein further inhibits eNOS, interfering with endothelial function. UPR-induced NF-κB activity regulates inflammatory processes in lung tissue and contributes to pulmonary vascular remodeling. Conversely, UPR-activated nuclear factor erythroid 2-related factor 2-mediated antioxidant signaling through heme oxygenase 1 attenuates inflammatory cytokine levels and protects against vascular SMC proliferation. A mutation in the bone morphogenic protein type 2 receptor (BMPR2) gene causes misfolded BMPR2 protein accumulation in the ER, implicating the UPR in familial pulmonary arterial hypertension pathogenesis. Altogether, there is substantial evidence that redox and inflammatory signaling associated with UPR activation is critical in PH pathogenesis.


Subject(s)
Hypertension, Pulmonary , Endoplasmic Reticulum Stress , Humans , Hypertension, Pulmonary/genetics , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Signal Transduction , Unfolded Protein Response
2.
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
4.
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
5.
Am J Physiol Regul Integr Comp Physiol ; 307(4): R426-33, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24920729

ABSTRACT

Exposing mice to a chronic hypoxic treatment (10% oxygen, 21 days) that promotes pulmonary hypertension was observed to attenuate the pulmonary vasoconstriction response to acute hypoxia (HPV) both in vivo and in isolated pulmonary arteries. Since catalase restored the HPV response in isolated arteries, it appeared to be attenuated by extracellular hydrogen peroxide. Chronic hypoxia promoted the detection of elevated lung superoxide, extracellular peroxide, extracellular SOD expression, and protein kinase G (PKG) activation [based on PKG dimerization and vasodilator-stimulated phosphoprotein (VASP) phosphorylation], suggesting increased generation of extracellular peroxide and PKG activation may contribute to the suppression of HPV. Aorta from mice exposed to 21 days of hypoxia also showed evidence for extracellular hydrogen peroxide, suppressing the relaxation response to acute hypoxia. Peroxide appeared to partially suppress contractions to phenylephrine used in the study of in vitro hypoxic responses. Treatment of mice with the heme precursor δ-aminolevulinic acid (ALA; 50 mg·kg(-1)·day(-1)) during exposure to chronic hypoxia was examined as a pulmonary hypertension therapy because it could potentially activate beneficial cGMP-mediated effects through promoting a prolonged protoporphyrin IX (PpIX)-elicited activation of soluble guanylate cyclase. ALA attenuated pulmonary hypertension, increases in both superoxide and peroxide, and the suppression of in vitro and in vivo HPV responses. ALA generated prolonged detectible increases in PpIX and PKG-associated phosphorylation of VASP, suggesting PKG activation may contribute to suppression of pulmonary hypertension and prevention of alterations in extracellular peroxide that appear to be attenuating HPV responses caused by chronic hypoxia.


Subject(s)
Hydrogen Peroxide/metabolism , Hypertension, Pulmonary/etiology , Hypoxia/complications , Pulmonary Artery/metabolism , Vasoconstriction , Acute Disease , Aminolevulinic Acid/pharmacology , Animals , Antihypertensive Agents/pharmacology , Aorta/metabolism , Aorta/physiopathology , Cell Adhesion Molecules/metabolism , Chronic Disease , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Enzyme Activation , Familial Primary Pulmonary Hypertension , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , Hypoxia/drug therapy , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protoporphyrins/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Superoxide Dismutase/metabolism , Superoxides/metabolism , Time Factors , Up-Regulation , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL