Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Mol Biosci ; 9: 989809, 2022.
Article in English | MEDLINE | ID: mdl-36275633

ABSTRACT

Acid-sensing ion channel 1a (ASIC1a) is a voltage-independent, non-selective cation channel that conducts both Na+ and Ca2+. Activation of ASIC1a elicits plasma membrane depolarization and stimulates intracellular Ca2+-dependent signaling pathways in multiple cell types, including vascular smooth muscle (SM) and endothelial cells (ECs). Previous studies have shown that increases in pulmonary vascular resistance accompanying chronic hypoxia (CH)-induced pulmonary hypertension requires ASIC1a to elicit enhanced pulmonary vasoconstriction and vascular remodeling. Both SM and EC dysfunction drive these processes; however, the involvement of ASIC1a within these different cell types is unknown. Using the Cre-LoxP system to generate cell-type-specific Asic1a knockout mice, we tested the hypothesis that SM-Asic1a contributes to CH-induced pulmonary hypertension and vascular remodeling, whereas EC-Asic1a opposes the development of CH-induced pulmonary hypertension. The severity of pulmonary hypertension was not altered in mice with specific deletion of EC-Asic1a (TekCre-Asic1a fl/fl). However, similar to global Asic1a knockout (Asic1a -/-) mice, mice with specific deletion of SM-Asic1a (MHCCreER-Asic1a fl/fl) were protected from the development of CH-induced pulmonary hypertension and right heart hypertrophy. Furthermore, pulmonary hypertension was reversed when deletion of SM-Asic1a was initiated in conditional MHCCreER-Asic1a fl/fl mice with established pulmonary hypertension. CH-induced vascular remodeling was also significantly attenuated in pulmonary arteries from MHCCreER-Asic1a fl/fl mice. These findings were additionally supported by decreased CH-induced proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) from Asic1a -/- mice. Together these data demonstrate that SM-, but not EC-Asic1a contributes to CH-induced pulmonary hypertension and vascular remodeling. Furthermore, these studies provide evidence for the therapeutic potential of ASIC1a inhibition to reverse pulmonary hypertension.

2.
PLoS One ; 15(7): e0236288, 2020.
Article in English | MEDLINE | ID: mdl-32702049

ABSTRACT

Although voltage-gated Ca2+ channels (VGCC) are a major Ca2+ entry pathway in vascular smooth muscle cells (VSMCs), several other Ca2+-influx mechanisms exist and play important roles in vasoreactivity. One of these is store-operated Ca2+ entry (SOCE), mediated by an interaction between STIM1 and Orai1. Although SOCE is an important mechanism of Ca2+ influx in non-excitable cells (cells that lack VGCC); there is debate regarding the contribution of SOCE to regulate VSMC contractility and the molecular components involved. Our previous data suggest acid-sensing ion channel 1a (ASIC1a) is a necessary component of SOCE and vasoconstriction in small pulmonary arteries. However, it is unclear if ASIC1a similarly contributes to SOCE and vascular reactivity in systemic arteries. Considering the established role of Orai1 in mediating SOCE in the systemic circulation, we hypothesize the involvement of ASIC1a in SOCE and resultant vasoconstriction is unique to the pulmonary circulation. To test this hypothesis, we examined the roles of Orai1 and ASIC1a in SOCE- and endothelin-1 (ET-1)-induced vasoconstriction in small pulmonary and mesenteric arteries. We found SOCE is coupled to vasoconstriction in pulmonary arteries but not mesenteric arteries. In pulmonary arteries, inhibition of ASIC1a but not Orai1 attenuated SOCE- and ET-1-induced vasoconstriction. However, neither inhibition of ASIC1a nor Orai1 altered ET-1-induced vasoconstriction in mesenteric arteries. We conclude that SOCE plays an important role in pulmonary, but not mesenteric, vascular reactivity. Furthermore, in contrast to the established role of Orai1 in SOCE in non-excitable cells, the SOCE response in pulmonary VSMCs is largely mediated by ASIC1a.


Subject(s)
Acid Sensing Ion Channels/metabolism , Calcium/metabolism , Mesenteric Arteries/physiology , Pulmonary Artery/physiology , Vasoconstriction , Acid Sensing Ion Channels/genetics , Animals , Calcium Channels, L-Type/metabolism , Endothelin-1/pharmacology , Male , Mesenteric Arteries/drug effects , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Protein Binding/drug effects , Pulmonary Artery/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Stromal Interaction Molecule 1/metabolism
3.
Am J Respir Cell Mol Biol ; 62(6): 732-746, 2020 06.
Article in English | MEDLINE | ID: mdl-32048876

ABSTRACT

Pulmonary vasoconstriction resulting from intermittent hypoxia (IH) contributes to pulmonary hypertension (pHTN) in patients with sleep apnea (SA), although the mechanisms involved remain poorly understood. Based on prior studies in patients with SA and animal models of SA, the objective of this study was to evaluate the role of PKCß and mitochondrial reactive oxygen species (mitoROS) in mediating enhanced pulmonary vasoconstrictor reactivity after IH. We hypothesized that PKCß mediates vasoconstriction through interaction with the scaffolding protein PICK1 (protein interacting with C kinase 1), activation of mitochondrial ATP-sensitive potassium channels (mitoKATP), and stimulated production of mitoROS. We further hypothesized that this signaling axis mediates enhanced vasoconstriction and pHTN after IH. Rats were exposed to IH or sham conditions (7 h/d, 4 wk). Chronic oral administration of the antioxidant Tempol or the PKCß inhibitor LY-333531 abolished IH-induced increases in right ventricular systolic pressure and right ventricular hypertrophy. Furthermore, scavengers of O2- or mitoROS prevented enhanced PKCß-dependent vasoconstrictor reactivity to endothelin-1 in pulmonary arteries from IH rats. In addition, this PKCß/mitoROS signaling pathway could be stimulated by the PKC activator PMA in pulmonary arteries from control rats, and in both rat and human pulmonary arterial smooth muscle cells. These responses to PMA were attenuated by inhibition of mitoKATP or PICK1. Subcellular fractionation and proximity ligation assays further demonstrated that PKCß acutely translocates to mitochondria upon stimulation and associates with PICK1. We conclude that a PKCß/mitoROS signaling axis contributes to enhanced vasoconstriction and pHTN after IH. Furthermore, PKCß mediates pulmonary vasoconstriction through interaction with PICK1, activation of mitoKATP, and subsequent mitoROS generation.


Subject(s)
Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Mitochondria/physiology , Protein Kinase C beta/physiology , Pulmonary Artery/physiopathology , Vasoconstriction/physiology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cells, Cultured , Cyclic N-Oxides/pharmacology , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/metabolism , Free Radical Scavengers/pharmacology , Humans , Hypertension, Pulmonary/etiology , Hypoxia/complications , Hypoxia/enzymology , Indoles/pharmacology , Male , Maleimides/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Potassium Channels/metabolism , Protein Interaction Mapping , Pulmonary Artery/enzymology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Signal Transduction , Sleep Apnea Syndromes/physiopathology , Spin Labels , Tetradecanoylphorbol Acetate/pharmacology
4.
J Appl Physiol (1985) ; 127(2): 393-407, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31169471

ABSTRACT

Acid-sensing ion channels (ASICs) are voltage-insensitive cation channels that contribute to cellular excitability. We previously reported that ASIC1 in pulmonary artery smooth muscle cells (PASMC) contribute to pulmonary vasoreactivity and vascular remodeling during the development of chronic hypoxia (CH)-induced pulmonary hypertension. However, the roles of ASIC2 and ASIC3 in regulation of pulmonary vasoreactivity and the development of CH-induced pulmonary hypertension are unknown. We tested the hypothesis that ASIC2 and ASIC3 contribute to increased pulmonary vasoreactivity and development of CH-induced pulmonary hypertension using ASIC2- and ASIC3-knockout (-/-) mice. In contrast to this hypothesis, we found that ASIC2-/- mice exhibit enhanced CH-induced pulmonary hypertension compared with WT and ASIC3-/- mice. This response was not associated with a change in ventilatory sensitivity or systemic cardiovascular function but was instead associated with direct changes in pulmonary vascular reactivity and pulmonary arterial morphology in ASIC2-/- mice. This increase in reactivity correlated with enhanced pulmonary arterial basal tone, elevated basal PASMC [Ca2+] and store-operated calcium entry (SOCE) in PASMC from ASIC2-/- mice. This increase in PASMC [Ca2+] and vasoreactivity was dependent on ASIC1-mediated Ca2+ influx but was not contingent upon an increase in ASIC1 mRNA or protein expression in PASMC from ASIC2-/- mice. Together, the results from this study demonstrate an important role for ASIC2 to regulate pulmonary vascular reactivity and for ASIC2 to modulate the development of CH-induced pulmonary hypertension. These data further suggest that loss of ASIC2 enhances the contribution of ASIC1 to overall pulmonary vascular reactivity.NEW & NOTEWORTHY This study demonstrates that loss of ASIC2 leads to increased baseline pulmonary vascular resistance, enhanced responses to a variety of vasoconstrictor stimuli, and greater development of hypoxic pulmonary hypertension. Furthermore, these results suggest that loss of ASIC2 enhances the contribution of ASIC1 to pulmonary vascular reactivity.


Subject(s)
Acid Sensing Ion Channels/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Lung/metabolism , Pulmonary Artery/metabolism , Vascular Resistance/physiology , Animals , Calcium/metabolism , Calcium Signaling/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Vascular Remodeling/physiology , Vasoconstriction/physiology
5.
Am J Physiol Cell Physiol ; 314(2): C166-C176, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29070491

ABSTRACT

Increases in pulmonary arterial smooth muscle cell (PASMC) intracellular Ca2+ levels and enhanced RhoA/Rho kinase-dependent Ca2+ sensitization are key determinants of PASMC contraction, migration, and proliferation accompanying the development of hypoxic pulmonary hypertension. We previously showed that acid-sensing ion channel 1a (ASIC1a)-mediated Ca2+ entry in PASMC is an important constituent of the active vasoconstriction, vascular remodeling, and right ventricular hypertrophy associated with hypoxic pulmonary hypertension. However, the enhanced ASIC1a-mediated store-operated Ca2+ entry in PASMC from pulmonary hypertensive animals is not dependent on an increase in ASIC1a protein expression, suggesting that chronic hypoxia (CH) stimulates ASIC1a function through other regulatory mechanism(s). RhoA is involved in ion channel trafficking, and levels of activated RhoA are increased following CH. Therefore, we hypothesize that activation of RhoA following CH increases ASIC1a-mediated Ca2+ entry by promoting ASIC1a plasma membrane localization. Consistent with our hypothesis, we found greater plasma membrane localization of ASIC1a following CH. Inhibition of RhoA decreased ASIC1a plasma membrane expression and largely diminished ASIC1a-mediated Ca2+ influx, whereas activation of RhoA had the opposite effect. A proximity ligation assay revealed that ASIC1a and RhoA colocalize in PASMC and that the activation state of RhoA modulates this interaction. Together, our findings show a novel interaction between RhoA and ASIC1a, such that activation of RhoA in PASMC, both pharmacologically and via CH, promotes ASIC1a plasma membrane localization and Ca2+ entry. In addition to enhanced RhoA-mediated Ca2+ sensitization following CH, RhoA can also activate a Ca2+ signal by facilitating ASIC1a plasma membrane localization and Ca2+ influx in pulmonary hypertension.


Subject(s)
Acid Sensing Ion Channels/metabolism , Calcium Signaling , Cell Membrane/enzymology , Hypertension, Pulmonary/enzymology , Hypoxia/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , rho GTP-Binding Proteins/metabolism , Acid Sensing Ion Channels/deficiency , Acid Sensing Ion Channels/genetics , Animals , Cell Membrane/pathology , Cells, Cultured , Chronic Disease , Disease Models, Animal , Female , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypoxia/genetics , Hypoxia/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Protein Transport , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Rats, Wistar , Time Factors , rhoA GTP-Binding Protein
6.
Inhal Toxicol ; 29(6): 266-281, 2017 05.
Article in English | MEDLINE | ID: mdl-28816559

ABSTRACT

Recent studies have shown a strong correlation between air pollution-exposure and detrimental outcomes in the central nervous system, including alterations in blood brain barrier (BBB) integrity, neuroinflammation, and neurodegeneration. However, the mechanisms mediating these pathologies have not yet been fully elucidated. We have previously reported that exposure to traffic-generated air pollution results in increased circulating oxidized low-density lipoprotein (oxLDL), associated with alterations in BBB integrity, in atherosclerotic Apolipoprotein E null (ApoE-/-) mice. Thus, we investigated the role of the lectin-like oxLDL receptor (LOX)-1 in mediating these deleterious effects in ApoE-/- mice exposed to a mixture of gasoline and diesel engine exhaust (MVE: 100 PM µg/m3) for 6 h/d, 7d/week, for 30 d by inhalation. Concurrent with exposures, a subset of mice were treated with neutralizing antibodies to LOX-1 (LOX-1 Ab) i.p., or IgG (control) i.p., every other day during exposures. Resulting brain microvascular integrity, tight junction (TJ) protein expression, matrix metalloproteinase (MMP)-9/-2 activity, ROS, and markers of cellular adhesion and monocyte/macrophage sequestration were assessed. MVE-exposure resulted in decreased BBB integrity and alterations in microvascular TJ protein expression, associated with increased LOX-1 expression, MMP-9/-2 activities, and lipid peroxidation, each of which was attenuated with LOX-1 Ab treatment. Furthermore, MVE-exposure induced cerebral microvascular ROS and adhesion molecules, expression of which was not normalized through LOX-1 Ab-treatment. Such findings suggest that alterations in brain microvascular structure and integrity observed with MVE-exposure may be mediated, at least in part, via LOX-1 signaling.


Subject(s)
Brain/drug effects , Microvessels/drug effects , Scavenger Receptors, Class E/metabolism , Vehicle Emissions/toxicity , Air Pollution/adverse effects , Animals , Antibodies, Neutralizing/pharmacology , Brain/blood supply , Brain/metabolism , Chemokine CCL2/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lipid Peroxidation/drug effects , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Knockout, ApoE , Microvessels/metabolism , Permeability , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class E/immunology , Vascular Cell Adhesion Molecule-1/metabolism
7.
PLoS One ; 12(6): e0180455, 2017.
Article in English | MEDLINE | ID: mdl-28666030

ABSTRACT

Pulmonary arterial hypertension is associated with a decreased antioxidant capacity. However, neither the contribution of reactive oxygen species to pulmonary vasoconstrictor sensitivity, nor the therapeutic efficacy of antioxidant strategies in this setting are known. We hypothesized that reactive oxygen species play a central role in mediating both vasoconstrictor and arterial remodeling components of severe pulmonary arterial hypertension. We examined the effect of the chemical antioxidant, TEMPOL, on right ventricular systolic pressure, vascular remodeling, and enhanced vasoconstrictor reactivity in both chronic hypoxia and hypoxia/SU5416 rat models of pulmonary hypertension. SU5416 is a vascular endothelial growth factor receptor antagonist and the combination of chronic hypoxia/SU5416 produces a model of severe pulmonary arterial hypertension with vascular plexiform lesions/fibrosis that is not present with chronic hypoxia alone. The major findings from this study are: 1) compared to hypoxia alone, hypoxia/SU5416 exposure caused more severe pulmonary hypertension, right ventricular hypertrophy, adventitial lesion formation, and greater vasoconstrictor sensitivity through a superoxide and Rho kinase-dependent Ca2+ sensitization mechanism. 2) Chronic hypoxia increased medial muscularization and superoxide levels, however there was no effect of SU5416 to augment these responses. 3) Treatment with TEMPOL decreased right ventricular systolic pressure in both hypoxia and hypoxia/SU5416 groups. 4) This effect of TEMPOL was associated with normalization of vasoconstrictor responses, but not arterial remodeling. Rather, medial hypertrophy and adventitial fibrotic lesion formation were more pronounced following chronic TEMPOL treatment in hypoxia/SU5416 rats. Our findings support a major role for reactive oxygen species in mediating enhanced vasoconstrictor reactivity and pulmonary hypertension in both chronic hypoxia and hypoxia/SU5416 rat models, despite a paradoxical effect of antioxidant therapy to exacerbate arterial remodeling in animals with severe pulmonary arterial hypertension in the hypoxia/SU5416 model.


Subject(s)
Disease Models, Animal , Hypertension, Pulmonary/metabolism , Pulmonary Artery/pathology , Reactive Oxygen Species/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Cyclic N-Oxides/administration & dosage , Endothelin-1/metabolism , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/pathology , Male , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Spin Labels , Superoxides/metabolism , Vasoconstriction
8.
Am J Physiol Lung Cell Mol Physiol ; 311(1): L48-58, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27190058

ABSTRACT

The development of chronic hypoxia (CH)-induced pulmonary hypertension is associated with increased pulmonary arterial smooth muscle cell (PASMC) Ca(2+) influx through acid-sensing ion channel-1 (ASIC1) and activation of the Ca(2+)/calcineurin-dependent transcription factor known as nuclear factor of activated T-cells isoform c3 (NFATc3). Whether Ca(2+) influx through ASIC1 contributes to NFATc3 activation in the pulmonary vasculature is unknown. Furthermore, both ASIC1 and calcineurin have been shown to interact with the scaffolding protein known as protein interacting with C kinase-1 (PICK1). In the present study, we tested the hypothesis that ASIC1 contributes to NFATc3 nuclear translocation in PASMC in a PICK1-dependent manner. Using both ASIC1 knockout (ASIC1(-/-)) mice and pharmacological inhibition of ASIC1, we demonstrate that ASIC1 contributes to CH-induced (1 wk at 380 mmHg) and endothelin-1 (ET-1)-induced (10(-7) M) Ca(2+) responses and NFATc3 nuclear import in PASMC. The interaction between ASIC1/PICK1/calcineurin was shown using a Duolink in situ Proximity Ligation Assay. Inhibition of PICK1 by using FSC231 abolished ET-1-induced and ionomycin-induced NFATc3 nuclear import, but it did not alter ET-1-mediated Ca(2+) responses, suggesting that PICK1 acts downstream of Ca(2+) influx. The key findings of the present work are that 1) Ca(2+) influx through ASIC1 mediates CH- and ET-1-induced NFATc3 nuclear import and 2) the scaffolding protein PICK1 is necessary for NFATc3 nuclear import. Together, these data provide an essential link between CH-induced ASIC1-mediated Ca(2+) influx and activation of the NFATc3 transcription factor. Identification of this ASIC1/PICK1/NFATc3 signaling complex increases our understanding of the mechanisms contributing to the vascular remodeling and increased vascular contractility that are associated with CH-induced pulmonary hypertension.


Subject(s)
Acid Sensing Ion Channels/physiology , Carrier Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , NFATC Transcription Factors/metabolism , Nuclear Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Calcium Signaling , Cell Cycle Proteins , Cell Hypoxia , Cells, Cultured , Endothelin-1/physiology , Female , Hypertension, Pulmonary/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/pathology
9.
Am J Physiol Cell Physiol ; 310(5): C390-400, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26702130

ABSTRACT

Acid-sensing ion channel 1 (ASIC1) contributes to Ca(2+) influx and contraction in pulmonary arterial smooth muscle cells (PASMC). ASIC1 binds the PDZ (PSD-95/Dlg/ZO-1) domain of the protein interacting with C kinase 1 (PICK1), and this interaction is important for the subcellular localization and/or activity of ASIC1. Therefore, we first hypothesized that PICK1 facilitates ASIC1-dependent Ca(2+) influx in PASMC by promoting plasma membrane localization. Using Duolink to determine protein-protein interactions and a biotinylation assay to assess membrane localization, we demonstrated that the PICK1 PDZ domain inhibitor FSC231 diminished the colocalization of PICK1 and ASIC1 but did not limit ASIC1 plasma membrane localization. Although stimulation of store-operated Ca(2+) entry (SOCE) greatly enhanced colocalization between ASIC1 and PICK1, both FSC231 and shRNA knockdown of PICK1 largely augmented SOCE. These data suggest PICK1 imparts a basal inhibitory effect on ASIC1 Ca(2+) entry in PASMC and led to an alternative hypothesis that PICK1 facilitates the interaction between ASIC1 and negative intracellular modulators, namely PKC and/or the calcium-calmodulin-activated phosphatase calcineurin. FSC231 limited PKC-mediated inhibition of SOCE, supporting a potential role for PICK1 in this response. Additionally, we found PICK1 inhibits ASIC1-mediated SOCE through an effect of calcineurin to dephosphorylate the channel. Furthermore, it appears PICK1/calcineurin-mediated regulation of SOCE opposes PKA phosphorylation and activation of ASIC1. Together our data suggest PKA and PICK1/calcineurin differentially regulate ASIC1-mediated SOCE and these modulatory complexes are important in determining downstream Ca(2+) signaling.


Subject(s)
Acid Sensing Ion Channels/drug effects , Calcineurin/metabolism , Calcium/metabolism , Carrier Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/metabolism , Acid Sensing Ion Channels/metabolism , Animals , Calcium Signaling/physiology , Cytoskeletal Proteins , Hypoxia/metabolism , Male , Pulmonary Artery/metabolism , Rats, Wistar
10.
Am J Physiol Lung Cell Mol Physiol ; 307(5): L419-30, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24993130

ABSTRACT

Our laboratory shows that acid-sensing ion channel 1 (ASIC1) contributes to the development of hypoxic pulmonary hypertension by augmenting store-operated Ca(2+) entry (SOCE) that is associated with enhanced agonist-induced vasoconstriction and arterial remodeling. However, this enhanced Ca(2+) influx following chronic hypoxia (CH) is not dependent on an increased ASIC1 protein expression in pulmonary arterial smooth muscle cells (PASMC). It is well documented that hypoxic pulmonary hypertension is associated with changes in redox potential and reactive oxygen species homeostasis. ASIC1 is a redox-sensitive channel showing increased activity in response to reducing agents, representing an alternative mechanism of regulation. We hypothesize that the enhanced SOCE following CH results from removal of an inhibitory effect of hydrogen peroxide (H2O2) on ASIC1. We found that CH increased PASMC superoxide (O2 (·-)) and decreased rat pulmonary arterial H2O2 levels. This decrease in H2O2 is a result of decreased Cu/Zn superoxide dismutase expression and activity, as well as increased glutathione peroxidase (GPx) expression and activity following CH. Whereas H2O2 inhibited ASIC1-dependent SOCE in PASMC from control and CH animals, addition of catalase augmented ASIC1-mediated SOCE in PASMC from control rats but had no further effect in PASMC from CH rats. These data suggest that, under control conditions, H2O2 inhibits ASIC1-dependent SOCE. Furthermore, H2O2 levels are decreased following CH as a result of diminished dismutation of O2 (·-) and increased H2O2 catalysis through GPx-1, leading to augmented ASIC1-dependent SOCE.


Subject(s)
Acid Sensing Ion Channels/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Hydrogen Peroxide/pharmacology , Hypoxia , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Animals , Blotting, Western , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Oxidants/pharmacology , Pulmonary Artery/cytology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/physiology , Superoxide Dismutase-1
11.
Am J Physiol Heart Circ Physiol ; 306(1): H41-52, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24186095

ABSTRACT

Chronic hypoxia (CH) associated with respiratory disease results in elevated pulmonary vascular intracellular Ca(2+) concentration, which elicits enhanced vasoconstriction and promotes vascular arterial remodeling and thus has important implications in the development of pulmonary hypertension (PH). Store-operated Ca(2+) entry (SOCE) contributes to this elevated intracellular Ca(2+) concentration and has also been linked to acute hypoxic pulmonary vasoconstriction (HPV). Since our laboratory has recently demonstrated an important role for acid-sensing ion channel 1 (ASIC1) in mediating SOCE, we hypothesized that ASIC1 contributes to both HPV and the development of CH-induced PH. To test this hypothesis, we examined responses to acute hypoxia in isolated lungs and assessed the effects of CH on indexes of PH, arterial remodeling, and vasoconstrictor reactivity in wild-type (ASIC1(+/+)) and ASIC1 knockout (ASIC1(-/-)) mice. Restoration of ASIC1 expression in pulmonary arterial smooth muscle cells from ASIC1(-/-) mice rescued SOCE, confirming the requirement for ASIC1 in this response. HPV responses were blunted in lungs from ASIC1(-/-) mice. Both SOCE and receptor-mediated Ca(2+) entry, along with agonist-dependent vasoconstrictor responses, were diminished in small pulmonary arteries from control ASIC(-/-) mice compared with ASIC(+/+) mice. The effects of CH to augment receptor-mediated vasoconstrictor and SOCE responses in vessels from ASIC1(+/+) mice were not observed after CH in ASIC1(-/-) mice. In addition, ASIC1(-/-) mice exhibited diminished right ventricular systolic pressure, right ventricular hypertrophy, and arterial remodeling in response to CH compared with ASIC1(+/+) mice. Taken together, these data demonstrate an important role for ASIC1 in both HPV and the development of CH-induced PH.


Subject(s)
Acid Sensing Ion Channels/genetics , Calcium Signaling , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Vasoconstriction , Acid Sensing Ion Channels/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Heart Ventricles/pathology , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Lung/blood supply , Lung/metabolism , Lung/physiopathology , Mice , Mice, Inbred C57BL , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Ventricular Dysfunction
12.
Part Fibre Toxicol ; 10: 62, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24344990

ABSTRACT

BACKGROUND: Traffic-generated air pollution-exposure is associated with adverse effects in the central nervous system (CNS) in both human exposures and animal models, including neuroinflammation and neurodegeneration. While alterations in the blood brain barrier (BBB) have been implicated as a potential mechanism of air pollution-induced CNS pathologies, pathways involved have not been elucidated. OBJECTIVES: To determine whether inhalation exposure to mixed vehicle exhaust (MVE) mediates alterations in BBB permeability, activation of matrix metalloproteinases (MMP) -2 and -9, and altered tight junction (TJ) protein expression. METHODS: Apolipoprotein (Apo) E(-/-) and C57Bl6 mice were exposed to either MVE (100 µg/m(3) PM) or filtered air (FA) for 6 hr/day for 30 days and resulting BBB permeability, expression of ROS, TJ proteins, markers of neuroinflammation, and MMP activity were assessed. Serum from study mice was applied to an in vitro BBB co-culture model and resulting alterations in transport and permeability were quantified. RESULTS: MVE-exposed Apo E(-/-) mice showed increased BBB permeability, elevated ROS and increased MMP-2 and -9 activity, compared to FA controls. Additionally, cerebral vessels from MVE-exposed mice expressed decreased levels of TJ proteins, occludin and claudin-5, and increased levels of inducible nitric oxide synthase (iNOS) and interleukin (IL)-1ß in the parenchyma. Serum from MVE-exposed animals also resulted in increased in vitro BBB permeability and altered P-glycoprotein transport activity. CONCLUSIONS: These data indicate that inhalation exposure to traffic-generated air pollutants promotes increased MMP activity and degradation of TJ proteins in the cerebral vasculature, resulting in altered BBB permeability and expression of neuroinflammatory markers.


Subject(s)
Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Inhalation Exposure/adverse effects , Matrix Metalloproteinases/genetics , Tight Junction Proteins/genetics , Vehicle Emissions/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/blood , Animals , Apolipoproteins E/genetics , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/metabolism , Blotting, Western , Coculture Techniques , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenic Inflammation/blood , Neurogenic Inflammation/chemically induced , Neurogenic Inflammation/enzymology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology
13.
Am J Physiol Cell Physiol ; 302(6): C931-40, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22205392

ABSTRACT

Acid-sensing ion channel 1 (ASIC1) is a newly characterized contributor to store-operated Ca(2+) entry (SOCE) in pulmonary vascular smooth muscle (VSM). Since SOCE is implicated in elevated basal VSM intracellular Ca(2+) concentration ([Ca(2+)](i)) and augmented vasoconstriction in chronic hypoxia (CH)-induced pulmonary hypertension, we hypothesized that ASIC1 contributes to these responses. To test this hypothesis, we examined effects of the specific pharmacologic ASIC1a inhibitor, psalmotoxin 1 (PcTX1), on vasoconstrictor and vessel wall [Ca(2+)](i) responses to UTP and KCl (depolarizing stimulus) in fura-2-loaded, pressurized small pulmonary arteries from control and CH (4 wk at 0.5 atm) Wistar rats. PcTX1 had no effect on basal vessel wall [Ca(2+)](i), but attenuated vasoconstriction and increases in vessel wall [Ca(2+)](i) to UTP in arteries from control and CH rats; normalizing responses between groups. In contrast, responses to the depolarizing stimulus, KCl, were unaffected by CH exposure or PcTX1. Upon examining potential Ca(2+) influx mechanisms, we found that PcTX1 prevented augmented SOCE following CH. Exposure to CH resulted in a significant increase in pulmonary arterial ASIC1 protein. This study supports a novel role of ASIC1 in elevated receptor-stimulated vasoconstriction following CH which is likely mediated through increased ASIC1 expression and SOCE.


Subject(s)
Calcium/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Nerve Tissue Proteins/metabolism , Pulmonary Artery/metabolism , Sodium Channels/metabolism , Acid Sensing Ion Channels , Animals , Calcium Channels, L-Type/metabolism , Fura-2/pharmacology , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Male , Muscle, Smooth, Vascular/blood supply , Muscle, Smooth, Vascular/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Peptides , Potassium Chloride/pharmacology , Pulmonary Artery/drug effects , Rats , Rats, Wistar , Spider Venoms/pharmacology , Uridine Triphosphate/pharmacology , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...