1
|
Lee SH, Ok SH, Park KE, Bae SI, Hwang Y, Ahn SH, Sim G, Bae M, Sohn JT. Epidermal growth factor receptor phosphorylation contributes to levobupivacaine-induced contraction in isolated rat aorta. Eur J Pharmacol 2024; 967:176389. [PMID: 38311282 DOI: 10.1016/j.ejphar.2024.176389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
Vasoconstriction induced by levobupivacaine, a local anesthetic, is mediated by increased levels of calcium, tyrosine kinase, c-Jun NH2-terminal kinase (JNK), and phospholipase D, which are associated with prolonged local anesthesia. Epidermal growth factor receptor (EGFR) phosphorylation is associated with vasoconstriction. However, its role in levobupivacaine-induced contractions remains unknown. We determined whether EGFR phosphorylation is associated with levobupivacaine-induced contractions in isolated rat thoracic aortas and identified the underlying cellular signaling pathways. The effects of various inhibitors and a calcium-free solution alone or in combination on levobupivacaine-induced contractions were then assessed. Furthermore, we examined the effects of various inhibitors on levobupivacaine-induced EGFR and JNK phosphorylation and calcium levels in vascular smooth muscle cells (VSMCs) of rat aortas. The EGFR tyrosine kinase inhibitor AG1478, matrix metalloproteinase (MMP) inhibitor GM6001, Src kinase inhibitors PP1 and PP2, and JNK inhibitor SP600125 attenuated levobupivacaine-induced contractions. Moreover, although the calcium-free solution abolished levobupivacaine-induced contractions, calcium reversed this inhibitory effect. The magnitude of the calcium-mediated reversal of abolished levobupivacaine-induced contractions was lower in the combination treatment with calcium-free solution and AG1478 than in the treatment with calcium-free solution alone. Levobupivacaine induced EGFR and JNK phosphorylation. However, AG1478, GM6001, and PP2 attenuated levobupivacaine-induced EGFR and JNK phosphorylation. Moreover, although levobupivacaine induced JNK phosphorylation in control siRNA-transfected VSMCs, EGFR siRNA inhibited levobupivacaine-induced JNK phosphorylation. Furthermore, AG1478 inhibited levobupivacaine-induced calcium increases in VSMCs. Collectively, these findings suggest that levobupivacaine-induced EGFR phosphorylation, which may occur via the Src kinase-MMP pathway, contributes to vasoconstriction via JNK phosphorylation and increased calcium levels.
Collapse
Affiliation(s)
- Soo Hee Lee
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Jinju-si, Gyeongsangnam-do, Republic of Korea; Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea
| | - Seong-Ho Ok
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Jinju-si, Gyeongsangnam-do, Republic of Korea; Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea
| | - Kyeong-Eon Park
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Sung Il Bae
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Yeran Hwang
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Seung Hyun Ahn
- Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Gyujin Sim
- Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Moonju Bae
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea
| | - Ju-Tae Sohn
- Institute of Medical Science, Gyeongsang National University, Jinju-si, Gyeongsangnam-do, Republic of Korea; Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Gyeongsang National University Hospital, 15 Jinju-daero 816 Beon-gil, Jinju-si, Gyeongsangnam-do, 52727, Republic of Korea.
| |
Collapse
|
2
|
Ji XY, Lei CJ, Kong S, Li HF, Pan SY, Chen YJ, Zhao FR, Zhu TT. Hydroxy-Safflower Yellow A Mitigates Vascular Remodeling in Rat Pulmonary Arterial Hypertension. Drug Des Devel Ther 2024; 18:475-491. [PMID: 38405578 PMCID: PMC10893878 DOI: 10.2147/dddt.s439686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose The underlying causes of pulmonary arterial hypertension (PAH) often remain obscure. Addressing PAH with effective treatments presents a formidable challenge. Studies have shown that Hydroxysafflor yellow A (HSYA) has a potential role in PAH, While the mechanism underlies its protective role is still unclear. The study was conducted to investigate the potential mechanisms of the protective effects of HSYA. Methods Using databases such as PharmMapper and GeneCards, we identified active components of HSYA and associated PAH targets, pinpointed intersecting genes, and constructed a protein-protein interaction (PPI) network. Core targets were singled out using Cytoscape for the development of a model illustrating drug-component-target-disease interactions. Intersection targets underwent analysis for Gene Ontology (GO) functions and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. Selected components were then modeled for target interaction using Autodock and Pymol. In vivo validation in a monocrotaline-induced PAH (MCT-PAH) animal model was utilized to substantiate the predictions made by network pharmacology. Results We associated HSYA with 113 targets, and PAH with 1737 targets, identifying 34 mutual targets for treatment by HSYA. HSYA predominantly affects 9 core targets. Molecular docking unveiled hydrogen bond interactions between HSYA and several PAH-related proteins such as ANXA5, EGFR, SRC, PPARG, PGR, and ESR1. Conclusion Utilizing network pharmacology and molecular docking approaches, we investigated potential targets and relevant human disease pathways implicating HSYA in PAH therapy, such as the chemical carcinogenesis receptor activation pathway and the cancer pathway. Our findings were corroborated by the efficacious use of HSYA in an MCT-induced rat PAH model, confirming its therapeutic potential.
Collapse
Affiliation(s)
- Xiang-Yu Ji
- Department of Pharmacy, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Cheng-Jing Lei
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Shuang Kong
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Han-Fei Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Si-Yu Pan
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Yu-Jing Chen
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Fan-Rong Zhao
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| | - Tian-Tian Zhu
- Department of Pharmacy, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, People’s Republic of China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, People’s Republic of China
| |
Collapse
|
3
|
Yan S, Sheak JR, Walker BR, Jernigan NL, Resta TC. Contribution of Mitochondrial Reactive Oxygen Species to Chronic Hypoxia-Induced Pulmonary Hypertension. Antioxidants (Basel) 2023; 12:2060. [PMID: 38136180 PMCID: PMC10741244 DOI: 10.3390/antiox12122060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Pulmonary hypertension (PH) resulting from chronic hypoxia (CH) occurs in patients with chronic obstructive pulmonary diseases, sleep apnea, and restrictive lung diseases, as well as in residents at high altitude. Previous studies from our group and others demonstrate a detrimental role of reactive oxygen species (ROS) in the pathogenesis of CH-induced PH, although the subcellular sources of ROS are not fully understood. We hypothesized that mitochondria-derived ROS (mtROS) contribute to enhanced vasoconstrictor reactivity and PH following CH. To test the hypothesis, we exposed rats to 4 weeks of hypobaric hypoxia (PB ≈ 380 mmHg), with control rats housed in ambient air (PB ≈ 630 mmHg). Chronic oral administration of the mitochondria-targeted antioxidant MitoQ attenuated CH-induced decreases in pulmonary artery (PA) acceleration time, increases in right ventricular systolic pressure, right ventricular hypertrophy, and pulmonary arterial remodeling. In addition, endothelium-intact PAs from CH rats exhibited a significantly greater basal tone compared to those from control animals, as was eliminated via MitoQ. CH also augmented the basal tone in endothelium-disrupted PAs, a response associated with increased mtROS production in primary PA smooth muscle cells (PASMCs) from CH rats. However, we further uncovered an effect of NO synthase inhibition with Nω-nitro-L-arginine (L-NNA) to unmask a potent endothelial vasoconstrictor influence that accentuates mtROS-dependent vasoconstriction following CH. This basal tone augmentation in the presence of L-NNA disappeared following combined endothelin A and B receptor blockade with BQ123 and BQ788. The effects of using CH to augment vasoconstriction and PASMC mtROS production in exogenous endothelin 1 (ET-1) were similarly prevented by MitoQ. We conclude that mtROS participate in the development of CH-induced PH. Furthermore, mtROS signaling in PASMCs is centrally involved in enhanced pulmonary arterial constriction following CH, a response potentiated by endogenous ET-1.
Collapse
Affiliation(s)
| | | | | | | | - Thomas C. Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA (J.R.S.); (B.R.W.); (N.L.J.)
| |
Collapse
|
4
|
Impact of Zinc on Oxidative Signaling Pathways in the Development of Pulmonary Vasoconstriction Induced by Hypobaric Hypoxia. Int J Mol Sci 2022; 23:ijms23136974. [PMID: 35805984 PMCID: PMC9266543 DOI: 10.3390/ijms23136974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 02/04/2023] Open
Abstract
Hypobaric hypoxia is a condition that occurs at high altitudes (>2500 m) where the partial pressure of gases, particularly oxygen (PO2), decreases. This condition triggers several physiological and molecular responses. One of the principal responses is pulmonary vascular contraction, which seeks to optimize gas exchange under this condition, known as hypoxic pulmonary vasoconstriction (HPV); however, when this physiological response is exacerbated, it contributes to the development of high-altitude pulmonary hypertension (HAPH). Increased levels of zinc (Zn2+) and oxidative stress (known as the “ROS hypothesis”) have been demonstrated in the vasoconstriction process. Therefore, the aim of this review is to determine the relationship between molecular pathways associated with altered Zn2+ levels and oxidative stress in HPV in hypobaric hypoxic conditions. The results indicate an increased level of Zn2+, which is related to increasing mitochondrial ROS (mtROS), alterations in nitric oxide (NO), metallothionein (MT), zinc-regulated, iron-regulated transporter-like protein (ZIP), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-induced protein kinase C epsilon (PKCε) activation in the development of HPV. In conclusion, there is an association between elevated Zn2+ levels and oxidative stress in HPV under different models of hypoxia, which contribute to understanding the molecular mechanism involved in HPV to prevent the development of HAPH.
Collapse
|
5
|
Norton CE, Shaw RL, Mittler R, Segal SS. Endothelial cells promote smooth muscle cell resilience to H 2 O 2 -induced cell death in mouse cerebral arteries. Acta Physiol (Oxf) 2022; 235:e13819. [PMID: 35380737 DOI: 10.1111/apha.13819] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 12/01/2022]
Abstract
AIM Brain injury produces reactive oxygen species (ROS). However, little is known of how acute oxidative stress affects cell survival in the cerebral vascular supply. We hypothesized that endothelial cells (ECs) are more resilient to H2 O2 and protect vascular smooth muscle cells (SMCs) during acute oxidative stress. METHODS Mouse posterior cerebral arteries (PCAs; diameter, ~80 µm) were exposed to H2 O2 (200 µM, 50 min, 37°C). Nuclear staining identified dead and live cells of intact and endothelium-disrupted vessels. SMC [Ca2+ ]i was assessed with Fura-2 fluorescence, and superoxide production was assessed by dihydroethidium and MitoSOX fluorescence. RESULTS In response to H2 O2 : SMC death (21%) exceeded EC death (5%) and increased following endothelial disruption (to 48%) with a corresponding increase in SMC Ca2+ entry through transient receptor potential (TRP) channels. Whereas pharmacological inhibition of TRPV4 channels prevented SMC death and reduced Ca2+ entry for intact vessels, both remained elevated following endothelial disruption. In contrast, pharmacological inhibition or genetic deletion of TRPC3 prevented SMC death and attenuated Ca2+ entry for both intact and endothelium-disrupted vessels. Inhibiting gap junctions increased EC death (to 22%) while SMC death and [Ca2+ ]i responses were attenuated by inhibiting nitric oxide synthesis or scavenging superoxide/peroxynitrite. Inhibiting NADPH oxidases also prevented SMC Ca2+ entry and death. H2 O2 increased mitochondrial ROS production while scavenging mitochondria-derived superoxide prevented SMC death but not Ca2+ entry. CONCLUSIONS During acute exposure of cerebral arteries to acute oxidative stress, ECs are more resilient than SMCs and the endothelium may protect SMCs by reducing Ca2+ entry through TRPC3 channels.
Collapse
Affiliation(s)
- Charles E. Norton
- Department of Medical Pharmacology and Physiology University of Missouri Columbia Missouri USA
| | - Rebecca L. Shaw
- Department of Medical Pharmacology and Physiology University of Missouri Columbia Missouri USA
| | - Ron Mittler
- Department of Surgery University of Missouri Columbia Missouri USA
| | - Steven S. Segal
- Department of Medical Pharmacology and Physiology University of Missouri Columbia Missouri USA
- Dalton Cardiovascular Research Center Columbia Missouri USA
| |
Collapse
|
6
|
Pena E, El Alam S, Siques P, Brito J. Oxidative Stress and Diseases Associated with High-Altitude Exposure. Antioxidants (Basel) 2022; 11:267. [PMID: 35204150 PMCID: PMC8868315 DOI: 10.3390/antiox11020267] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/12/2022] Open
Abstract
Several diseases associated with high-altitude exposure affect unacclimated individuals. These diseases include acute mountain sickness (AMS), high-altitude cerebral edema (HACE), high-altitude pulmonary edema (HAPE), chronic mountain sickness (CMS), and, notably, high-altitude pulmonary hypertension (HAPH), which can eventually lead to right ventricle hypertrophy and heart failure. The development of these pathologies involves different molecules and molecular pathways that might be related to oxidative stress. Studies have shown that acute, intermittent, and chronic exposure to hypobaric hypoxia induce oxidative stress, causing alterations to molecular pathways and cellular components (lipids, proteins, and DNA). Therefore, the aim of this review is to discuss the oxidative molecules and pathways involved in the development of high-altitude diseases. In summary, all high-altitude pathologies are related to oxidative stress, as indicated by increases in the malondialdehyde (MDA) biomarker and decreases in superoxide dismutase (SOD) and glutathione peroxidase (GPx) antioxidant activity. In addition, in CMS, the levels of 8-iso-PGF2α and H2O2 are increased, and evidence strongly indicates an increase in Nox4 activity in HAPH. Therefore, antioxidant treatments seem to be a promising approach to mitigating high-altitude pathologies.
Collapse
Affiliation(s)
- Eduardo Pena
- Institute of Health Studies, Arturo Prat University, Iquique 1100000, Chile; (E.P.); (P.S.); (J.B.)
| | - Samia El Alam
- Institute of Health Studies, Arturo Prat University, Iquique 1100000, Chile; (E.P.); (P.S.); (J.B.)
| | - Patricia Siques
- Institute of Health Studies, Arturo Prat University, Iquique 1100000, Chile; (E.P.); (P.S.); (J.B.)
| | - Julio Brito
- Institute of Health Studies, Arturo Prat University, Iquique 1100000, Chile; (E.P.); (P.S.); (J.B.)
| |
Collapse
|
7
|
Siques P, Pena E, Brito J, El Alam S. Oxidative Stress, Kinase Activation, and Inflammatory Pathways Involved in Effects on Smooth Muscle Cells During Pulmonary Artery Hypertension Under Hypobaric Hypoxia Exposure. Front Physiol 2021; 12:690341. [PMID: 34434114 PMCID: PMC8381601 DOI: 10.3389/fphys.2021.690341] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
High-altitude exposure results in hypobaric hypoxia, which affects organisms by activating several mechanisms at the physiological, cellular, and molecular levels and triggering the development of several pathologies. One such pathology is high-altitude pulmonary hypertension (HAPH), which is initiated through hypoxic pulmonary vasoconstriction to distribute blood to more adequately ventilated areas of the lungs. Importantly, all layers of the pulmonary artery (adventitia, smooth muscle, and endothelium) contribute to or are involved in the development of HAPH. However, the principal action sites of HAPH are pulmonary artery smooth muscle cells (PASMCs), which interact with several extracellular and intracellular molecules and participate in mechanisms leading to proliferation, apoptosis, and fibrosis. This review summarizes the alterations in molecular pathways related to oxidative stress, inflammation, kinase activation, and other processes that occur in PASMCs during pulmonary hypertension under hypobaric hypoxia and proposes updates to pharmacological treatments to mitigate the pathological changes in PASMCs under such conditions. In general, PASMCs exposed to hypobaric hypoxia undergo oxidative stress mediated by Nox4, inflammation mediated by increases in interleukin-6 levels and inflammatory cell infiltration, and activation of the protein kinase ERK1/2, which lead to the proliferation of PASMCs and contribute to the development of hypobaric hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Patricia Siques
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Eduardo Pena
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Julio Brito
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Samia El Alam
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| |
Collapse
|
8
|
Norton CE, Jernigan NL, Walker BR, Resta TC. Membrane depolarization is required for pressure-dependent pulmonary arterial tone but not enhanced vasoconstriction to endothelin-1 following chronic hypoxia. Pulm Circ 2020; 10:2045894020973559. [PMID: 33343882 PMCID: PMC7731711 DOI: 10.1177/2045894020973559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/23/2020] [Indexed: 11/28/2022] Open
Abstract
Enhanced vasoconstriction is increasingly identified as an important contributor to the development of pulmonary hypertension. Chronic hypoxia results in enhanced Rho kinase mediated Ca2+ sensitization contributing to pressure-dependent pulmonary arterial tone as well as augmented vasoconstriction to endothelin-1 and depolarizing stimuli. We sought to investigate the interaction between these vasoconstrictor stimuli in isolated, pressurized, pulmonary arteries. We used the K+ ionophore, valinomycin, to clamp membrane potential (Vm) to investigate the role of membrane depolarization in endothelin-1 and pressure-dependent constriction, and endothelin-1 receptor inhibitors to determine whether membrane depolarization or stretch signal through endothelin-1 receptors. Clamping Vm prevented pressure-dependent tone, but not enhanced vasoconstriction to endothelin-1 following chronic hypoxia. Furthermore, endothelin-1 receptor inhibition had no effect on either pressure-dependent tone or vasoconstriction to KCl. As Src kinases contribute to both pressure-dependent tone and enhanced endothelin-1 vasoconstriction following chronic hypoxia, we further investigated their role in depolarization-induced vasoconstriction. Inhibition of Src kinases attenuated enhanced vasoconstriction to KCl. We conclude that membrane depolarization contributes to pressure-dependent tone but not enhanced vasoconstriction to ET-1, and that Src kinases serve as upstream mediators facilitating enhanced Rho kinase-dependent vasoconstriction following chronic hypoxia.
Collapse
Affiliation(s)
- Charles E Norton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
9
|
Jin F, Guo Y, Wang Z, Badughaish A, Pan X, Zhang L, Qi F. The pathophysiological nature of sarcomeres in trigger points in patients with myofascial pain syndrome: A preliminary study. Eur J Pain 2020; 24:1968-1978. [PMID: 32841448 PMCID: PMC7693045 DOI: 10.1002/ejp.1647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/08/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Myofascial pain syndrome (MPS) has a high global prevalence and is associated with myofascial trigger points (MTrPs) in taut bands or nodules. Little is known about the aetiology. The current study assessed the pathophysiological characteristics of MTrPs in MPS patients. METHODS Biopsies of the trapezius muscle were collected from the MTrPs of MPS patients (MTrP group; n = 29) and from healthy controls (control group; n = 24), and their morphologies were analysed via haematoxylin-eosin (H&E) and Masson staining. A protein microarray was used to detect the receptor tyrosine kinase (RTK) family proteins. mRNA and long non-coding RNA (lncRNA) sequencing and analysis were conducted, and immunohistochemistry and Western blotting were used to examine the expression of EphB and Rho family proteins. RESULTS Abnormally contracted sarcomeres showed enlarged, round fibres without inflammation or fibrosis. An lncRNA-mRNA network analysis revealed activation of muscle contraction signalling pathways in MTrP regions. Among RTK family proteins, 15 exhibited increased phosphorylation, and two exhibited decreased phosphorylation in the MTrP regions relative to control levels. In particular, EphB1/EphB2 phosphorylation was increased on the muscle cell membranes of abnormal sarcomeres. RhoA and Rac1, but not cell division control protein 42 (Cdc42), were activated in the abnormal sarcomeres. CONCLUSIONS EphB1/EphB2 and RhoA/Rac1 might play roles in the aetiology of abnormally contracted sarcomeres in MTrPs without inflammatory cell infiltration and fibrotic adhesion. SIGNIFICANCE Contracted sarcomeres were found in MTrP regions, which is consistent with the MTrP formation hypothesis. EphB1/EphB2 and RhoA/Rac1 might play roles in the sarcomere contractile sites of MTrPs, which may be promising therapeutic targets.
Collapse
Affiliation(s)
- Feihong Jin
- Department of Anesthesiology and Pain ClinicQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJi’nanChina
| | - Yaqiu Guo
- Department of Anesthesiology and Pain ClinicQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJi’nanChina
- Department of AnesthesiologyJinan Maternity and Child Care HospitalJi’nanChina
| | - Zi Wang
- Department of Anesthesiology and Pain ClinicQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJi’nanChina
- Department of AnesthesiologyFirst Affiliated Hospital of Shandong TCM UniversityJi’nanChina
| | - Ahmed Badughaish
- Department of Anesthesiology and Pain ClinicQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJi’nanChina
| | - Xin Pan
- Department of OrthopedicsQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJi’nanChina
| | - Li Zhang
- Department of OrthopedicsQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJi’nanChina
| | - Feng Qi
- Department of Anesthesiology and Pain ClinicQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJi’nanChina
| |
Collapse
|
10
|
Yan S, Resta TC, Jernigan NL. Vasoconstrictor Mechanisms in Chronic Hypoxia-Induced Pulmonary Hypertension: Role of Oxidant Signaling. Antioxidants (Basel) 2020; 9:E999. [PMID: 33076504 PMCID: PMC7602539 DOI: 10.3390/antiox9100999] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
Elevated resistance of pulmonary circulation after chronic hypoxia exposure leads to pulmonary hypertension. Contributing to this pathological process is enhanced pulmonary vasoconstriction through both calcium-dependent and calcium sensitization mechanisms. Reactive oxygen species (ROS), as a result of increased enzymatic production and/or decreased scavenging, participate in augmentation of pulmonary arterial constriction by potentiating calcium influx as well as activation of myofilament sensitization, therefore mediating the development of pulmonary hypertension. Here, we review the effects of chronic hypoxia on sources of ROS within the pulmonary vasculature including NADPH oxidases, mitochondria, uncoupled endothelial nitric oxide synthase, xanthine oxidase, monoamine oxidases and dysfunctional superoxide dismutases. We also summarize the ROS-induced functional alterations of various Ca2+ and K+ channels involved in regulating Ca2+ influx, and of Rho kinase that is responsible for myofilament Ca2+ sensitivity. A variety of antioxidants have been shown to have beneficial therapeutic effects in animal models of pulmonary hypertension, supporting the role of ROS in the development of pulmonary hypertension. A better understanding of the mechanisms by which ROS enhance vasoconstriction will be useful in evaluating the efficacy of antioxidants for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
| | | | - Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.Y.); (T.C.R.)
| |
Collapse
|
11
|
Snow JB, Norton CE, Sands MA, Weise-Cross L, Yan S, Herbert LM, Sheak JR, Gonzalez Bosc LV, Walker BR, Kanagy NL, Jernigan NL, Resta TC. Intermittent Hypoxia Augments Pulmonary Vasoconstrictor Reactivity through PKCβ/Mitochondrial Oxidant Signaling. Am J Respir Cell Mol Biol 2020; 62:732-746. [PMID: 32048876 DOI: 10.1165/rcmb.2019-0351oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
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 PKCβ and mitochondrial reactive oxygen species (mitoROS) in mediating enhanced pulmonary vasoconstrictor reactivity after IH. We hypothesized that PKCβ 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 PKCβ inhibitor LY-333531 abolished IH-induced increases in right ventricular systolic pressure and right ventricular hypertrophy. Furthermore, scavengers of O2- or mitoROS prevented enhanced PKCβ-dependent vasoconstrictor reactivity to endothelin-1 in pulmonary arteries from IH rats. In addition, this PKCβ/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 PKCβ acutely translocates to mitochondria upon stimulation and associates with PICK1. We conclude that a PKCβ/mitoROS signaling axis contributes to enhanced vasoconstriction and pHTN after IH. Furthermore, PKCβ mediates pulmonary vasoconstriction through interaction with PICK1, activation of mitoKATP, and subsequent mitoROS generation.
Collapse
Affiliation(s)
- Jessica B Snow
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Charles E Norton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Michelle A Sands
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Simin Yan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Lindsay M Herbert
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Joshua R Sheak
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nancy L Kanagy
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
12
|
Grimmer B, Kuebler WM. Cholesterol: A Novel Regulator of Vasoreactivity in Pulmonary Arteries. Am J Respir Cell Mol Biol 2020; 62:671-673. [PMID: 32011912 PMCID: PMC7258827 DOI: 10.1165/rcmb.2020-0020ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Benjamin Grimmer
- Institute of PhysiologyCharité-University Medicine BerlinBerlin, Germany
| | - Wolfgang M Kuebler
- Institute of PhysiologyCharité-University Medicine BerlinBerlin, Germany.,The Keenan Research Centre for Biomedical ScienceSt. Michael's HospitalToronto, Ontario, Canada.,Department of Surgeryand.,Department of PhysiologyUniversity of TorontoToronto, Ontario, Canada
| |
Collapse
|
13
|
Sheak JR, Yan S, Weise-Cross L, Ahmadian R, Walker BR, Jernigan NL, Resta TC. PKCβ and reactive oxygen species mediate enhanced pulmonary vasoconstrictor reactivity following chronic hypoxia in neonatal rats. Am J Physiol Heart Circ Physiol 2020; 318:H470-H483. [PMID: 31922892 DOI: 10.1152/ajpheart.00629.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Reactive oxygen species (ROS), mitochondrial dysfunction, and excessive vasoconstriction are important contributors to chronic hypoxia (CH)-induced neonatal pulmonary hypertension. On the basis of evidence that PKCβ and mitochondrial oxidative stress are involved in several cardiovascular and metabolic disorders, we hypothesized that PKCβ and mitochondrial ROS (mitoROS) signaling contribute to enhanced pulmonary vasoconstriction in neonatal rats exposed to CH. To test this hypothesis, we examined effects of the PKCβ inhibitor LY-333,531, the ROS scavenger 1-oxyl-2,2,6,6-tetramethyl-4-hydroxypiperidine (TEMPOL), and the mitochondrial antioxidants mitoquinone mesylate (MitoQ) and (2-(2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (MitoTEMPO) on vasoconstrictor responses in saline-perfused lungs (in situ) or pressurized pulmonary arteries from 2-wk-old control and CH (12-day exposure, 0.5 atm) rats. Lungs from CH rats exhibited greater basal tone and vasoconstrictor sensitivity to 9,11-dideoxy-9α,11α-methanoepoxy prostaglandin F2α (U-46619). LY-333,531 and TEMPOL attenuated these effects of CH, while having no effect in lungs from control animals. Basal tone was similarly elevated in isolated pulmonary arteries from neonatal CH rats compared with control rats, which was inhibited by both LY-333,531 and mitochondria-targeted antioxidants. Additional experiments assessing mitoROS generation with the mitochondria-targeted ROS indicator MitoSOX revealed that a PKCβ-mitochondrial oxidant signaling pathway can be pharmacologically stimulated by the PKC activator phorbol 12-myristate 13-acetate in primary cultures of pulmonary artery smooth muscle cells (PASMCs) from control neonates. Finally, we found that neonatal CH increased mitochondrially localized PKCβ in pulmonary arteries as assessed by Western blotting of subcellular fractions. We conclude that PKCβ activation leads to mitoROS production in PASMCs from neonatal rats. Furthermore, this signaling axis may account for enhanced pulmonary vasoconstrictor sensitivity following CH exposure.NEW & NOTEWORTHY This research demonstrates a novel contribution of PKCβ and mitochondrial reactive oxygen species signaling to increased pulmonary vasoconstrictor reactivity in chronically hypoxic neonates. The results provide a potential mechanism by which chronic hypoxia increases both basal and agonist-induced pulmonary arterial smooth muscle tone, which may contribute to neonatal pulmonary hypertension.
Collapse
Affiliation(s)
- Joshua R Sheak
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Simin Yan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Rosstin Ahmadian
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
14
|
Norton CE, Weise-Cross L, Ahmadian R, Yan S, Jernigan NL, Paffett ML, Naik JS, Walker BR, Resta TC. Altered Lipid Domains Facilitate Enhanced Pulmonary Vasoconstriction after Chronic Hypoxia. Am J Respir Cell Mol Biol 2020; 62:709-718. [PMID: 31945301 DOI: 10.1165/rcmb.2018-0318oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic hypoxia (CH) augments depolarization-induced pulmonary vasoconstriction through superoxide-dependent, Rho kinase-mediated Ca2+ sensitization. Nicotinamide adenine dinucleotide phosphate oxidase and EGFR (epidermal growth factor receptor) signaling contributes to this response. Caveolin-1 regulates the activity of a variety of proteins, including EGFR and nicotinamide adenine dinucleotide phosphate oxidase, and membrane cholesterol is an important regulator of caveolin-1 protein interactions. We hypothesized that derangement of these membrane lipid domain components augments depolarization-induced Ca2+ sensitization and resultant vasoconstriction after CH. Although exposure of rats to CH (4 wk, ∼380 mm Hg) did not alter caveolin-1 expression in intrapulmonary arteries or the incidence of caveolae in arterial smooth muscle, CH markedly reduced smooth muscle membrane cholesterol content as assessed by filipin fluorescence. Effects of CH on vasoreactivity and superoxide generation were examined using pressurized, Ca2+-permeabilized, endothelium-disrupted pulmonary arteries (∼150 μm inner diameter) from CH and control rats. Depolarizing concentrations of KCl evoked greater constriction in arteries from CH rats than in those obtained from control rats, and increased superoxide production as assessed by dihydroethidium fluorescence only in arteries from CH rats. Both cholesterol supplementation and the caveolin-1 scaffolding domain peptide antennapedia-Cav prevented these effects of CH, with each treatment restoring membrane cholesterol in CH arteries to control levels. Enhanced EGF-dependent vasoconstriction after CH similarly required reduced membrane cholesterol. However, these responses to CH were not associated with changes in EGFR expression or activity, suggesting that cholesterol regulates this signaling pathway downstream of EGFR. We conclude that alterations in membrane lipid domain signaling resulting from reduced cholesterol content facilitate enhanced depolarization- and EGF-induced pulmonary vasoconstriction after CH.
Collapse
Affiliation(s)
- Charles E Norton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Rosstin Ahmadian
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Simin Yan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Michael L Paffett
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
15
|
Norton CE, Sheak JR, Yan S, Weise-Cross L, Jernigan NL, Walker BR, Resta TC. Augmented Pulmonary Vasoconstrictor Reactivity after Chronic Hypoxia Requires Src Kinase and Epidermal Growth Factor Receptor Signaling. Am J Respir Cell Mol Biol 2020; 62:61-73. [PMID: 31264901 PMCID: PMC6938133 DOI: 10.1165/rcmb.2018-0106oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/28/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic hypoxia augments pressure- and agonist-induced pulmonary vasoconstriction through myofilament calcium sensitization. NADPH oxidases contribute to the development of pulmonary hypertension, and both epidermal growth factor receptor and Src kinases can regulate NADPH oxidase. We tested the hypothesis that Src-epidermal growth factor receptor (EGFR) signaling mediates enhanced vasoconstrictor sensitivity after chronic hypoxia through NADPH oxidase-derived superoxide generation. Protocols employed pharmacological inhibitors in isolated, pressurized rat pulmonary arteries to examine the contribution of a variety of signaling moieties to enhanced vascular tone after chronic hypoxia. Superoxide generation in pulmonary arterial smooth muscle cells was assessed using the fluorescent indicator dihydroethidium. Indices of pulmonary hypertension were measured in rats treated with the EGFR inhibitor gefitinib. Inhibition of NADPH oxidase, Rac1 (Ras-related C3 botulinum toxin substrate 1), and EGFR abolished pressure-induced pulmonary arterial tone and endothelin-1 (ET-1)-dependent calcium sensitization and vasoconstriction after chronic hypoxia. Consistently, chronic hypoxia augmented ET-1-induced superoxide production through EGFR signaling, and rats treated chronically with gefitinib displayed reduced right ventricular pressure and diminished arterial remodeling. Src kinases were also activated by ET-1 after chronic hypoxia and contributed to enhanced basal arterial tone and vasoconstriction in response to ET-1. A role for matrix metalloproteinase 2 to mediate Src-dependent EGFR activation is further supported by our findings. Our studies support a novel role for an Src kinase-EGFR-NADPH oxidase signaling axis to mediate enhanced pulmonary vascular smooth muscle Ca2+ sensitization, vasoconstriction, and pulmonary hypertension after chronic hypoxia.
Collapse
Affiliation(s)
- Charles E Norton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Joshua R Sheak
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Simin Yan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
16
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
17
|
Weise-Cross L, Resta TC, Jernigan NL. Redox Regulation of Ion Channels and Receptors in Pulmonary Hypertension. Antioxid Redox Signal 2019; 31:898-915. [PMID: 30569735 PMCID: PMC7061297 DOI: 10.1089/ars.2018.7699] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 12/11/2018] [Indexed: 02/06/2023]
Abstract
Significance: Pulmonary hypertension (PH) is characterized by elevated vascular resistance due to vasoconstriction and remodeling of the normally low-pressure pulmonary vasculature. Redox stress contributes to the pathophysiology of this disease by altering the regulation and activity of membrane receptors, K+ channels, and intracellular Ca2+ homeostasis. Recent Advances: Antioxidant therapies have had limited success in treating PH, leading to a growing appreciation that reductive stress, in addition to oxidative stress, plays a role in metabolic and cell signaling dysfunction in pulmonary vascular cells. Reactive oxygen species generation from mitochondria and NADPH oxidases has substantial effects on K+ conductance and membrane potential, and both receptor-operated and store-operated Ca2+ entry. Critical Issues: Some specific redox changes resulting from oxidation, S-nitrosylation, and S-glutathionylation are known to modulate membrane receptor and ion channel activity in PH. However, many sites of regulation that have been elucidated in nonpulmonary cell types have not been tested in the pulmonary vasculature, and context-specific molecular mechanisms are lacking. Future Directions: Here, we review what is known about redox regulation of membrane receptors and ion channels in PH. Further investigation of the mechanisms involved is needed to better understand the etiology of PH and develop better targeted treatment strategies.
Collapse
Affiliation(s)
- Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C. Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
18
|
Gao Y, Raj JU. Src and Epidermal Growth Factor Receptor: Novel Partners in Mediating Chronic Hypoxia-induced Pulmonary Artery Hypertension. Am J Respir Cell Mol Biol 2019; 62:5-7. [PMID: 31298924 PMCID: PMC6938126 DOI: 10.1165/rcmb.2019-0230ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Yuansheng Gao
- Health Science CenterPeking UniversityBeijing, Chinaand
| | - J Usha Raj
- College of MedicineUniversity of Illinois at ChicagoChicago, Illinois
| |
Collapse
|
19
|
Kitagawa MG, Reynolds JO, Durgan D, Rodney G, Karmouty‐Quintana H, Bryan R, Pandit LM. Twik-2 -/- mouse demonstrates pulmonary vascular heterogeneity in intracellular pathways for vasocontractility. Physiol Rep 2019; 7:e13950. [PMID: 30632293 PMCID: PMC6328926 DOI: 10.14814/phy2.13950] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/06/2018] [Accepted: 11/15/2018] [Indexed: 11/24/2022] Open
Abstract
We have previously shown Twik-2-/- mice develop pulmonary hypertension and vascular remodeling. We hypothesized that distal pulmonary arteries (D-PAs) of the Twik-2-/- mice are hypercontractile under physiological venous conditions due to altered electrophysiologic properties between the conduit and resistance vessels in the pulmonary vascular bed. We measured resting membrane potential and intracellular calcium through Fura-2 in freshly digested pulmonary artery smooth muscles (PASMCs) from both the right main (RM-PA) and D-PA (distal) regions of pulmonary artery from WT and Twik-2-/- mice. Whole segments of RM-PAs and D-PAs from 20 to 24-week-old wildtype (WT) and Twik-2-/- mice were also pressurized between two glass micropipettes and bathed in buffer with either arterial or venous conditions. Abluminally-applied phenylephrine (PE) and U46619 were added to the buffer at log increments and vessel diameter was measured. All values were expressed as averages with ±SEM. Vasoconstrictor responses did not differ between WT and Twik-2-/- RM-PAs under arterial conditions. Under venous conditions, Twik-2-/- RM-PAs showed an increased sensitivity to PE with a lower EC50 (P = 0.02). Under venous conditions, Twik-2-/- D-PAs showed an increase maximal vasoconstrictor response to both phenylephrine and U46619 compared to the WT mice (P < 0.05). Isolated PASMCs from Twik-2 -/- D-PA were depolarized and had higher intracellular calcium levels compared to PASMCs from RM-PA of both WT and Twik-2-/- mice. These studies suggest that hypercontractile responses and electrophysiologic properties unique to the anatomic location of the D-PAs may contribute to pulmonary hypertensive vasculopathy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lavannya M. Pandit
- Baylor College of MedicineHoustonTexas
- Michael E.DeBakey Veterans Affairs Medical CenterHoustonTexas
| |
Collapse
|
20
|
Strassheim D, Karoor V, Stenmark K, Verin A, Gerasimovskaya E. A current view of G protein-coupled receptor - mediated signaling in pulmonary hypertension: finding opportunities for therapeutic intervention. ACTA ACUST UNITED AC 2018; 2. [PMID: 31380505 PMCID: PMC6677404 DOI: 10.20517/2574-1209.2018.44] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathological vascular remodeling is observed in various cardiovascular diseases including pulmonary hypertension (PH), a disease of unknown etiology that has been characterized by pulmonary artery vasoconstriction, right ventricular hypertrophy, vascular inflammation, and abnormal angiogenesis in pulmonary circulation. G protein-coupled receptors (GPCRs) are the largest family in the genome and widely expressed in cardiovascular system. They regulate all aspects of PH pathophysiology and represent therapeutic targets. We overview GPCRs function in vasoconstriction, vasodilation, vascular inflammation-driven remodeling and describe signaling cross talk between GPCR, inflammatory cytokines, and growth factors. Overall, the goal of this review is to emphasize the importance of GPCRs as critical signal transducers and targets for drug development in PH.
Collapse
Affiliation(s)
- Derek Strassheim
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Vijaya Karoor
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.,Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA
| | - Kurt Stenmark
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Evgenia Gerasimovskaya
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| |
Collapse
|
21
|
Moraga FA, Miranda G, López V, Vallejos C, Silva D. Chronic Intermittent Hypobaric Hypoxia (4600 M) Attenuates Pulmonary Vasodilation Induced by Acetylcholine or Sodium Nitroprusside. High Alt Med Biol 2018; 19:149-155. [PMID: 29565678 DOI: 10.1089/ham.2017.0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Moraga, Fernando A., Giselle Miranda, Vasthi López, Carmen Vallejos, and Daniel Silva. Chronic intermittent hypobaric hypoxia (4600 M) attenuates pulmonary vasodilation induced by acetylcholine or sodium nitroprusside. High Alt Med Biol. 19:149-155, 2018. BACKGROUND Previous studies performed in rats exposed to chronic intermittent hypobaric hypoxia (CIHH), at a simulated altitude of 4600 m, showed reduced nitric oxide (NO) production, increased arginase activity, and increased oxidative stress. However, studies on vascular function are scarce. Our aim was to measure plasma nitrate and nitrite (NOx) concentration and study pulmonary vascular function in rats exposed to CIHH in the presence of potassium chloride (KCl), acetylcholine (Ach), and sodium nitroprusside (SNP). METHODS Thirty male Wistar rats were divided into two groups: A control group (normoxia (N), n = 10) and a CIHH group (2N × 2H × 30 days, n = 20). CIHH exposure was performed in a hypobaric chamber at 428 Torr (4600 m). Noninvasive systolic blood pressure (SBP), heart rate, and body weight (BW) were measured. Blood samples were obtained to measure NOx levels and hematocrit (Hct). CIHH animals that gained BW and presented a Hct <20% and maintained SBP were classified as tolerant, and animals that lost >30% of their BW, increased Hct and SBP >20% were classified as intolerant. Animals were sacrificed and small pulmonary arteries (SPA) were obtained to perform concentration-response curves to KCl, Ach, and SNP. RESULTS AND CONCLUSIONS Intolerant rats (30%) had decreased NOx levels. SPA had a larger vasocontraction response to KCl and a lower dilation response to SNP in the SPA compared to tolerant and control animals. In addition, SPA had a lower dilatation response to Ach compared with the control. Together, these results show that CIHH alters endothelium-dependent vasodilation.
Collapse
Affiliation(s)
- Fernando A Moraga
- Laboratory of Physiology, Hypoxia and Vascular Function, Department of Biomedical Science, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, Chile
| | - Giselle Miranda
- Laboratory of Physiology, Hypoxia and Vascular Function, Department of Biomedical Science, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, Chile
| | - Vasthi López
- Laboratory of Physiology, Hypoxia and Vascular Function, Department of Biomedical Science, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, Chile
| | - Carmen Vallejos
- Laboratory of Physiology, Hypoxia and Vascular Function, Department of Biomedical Science, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, Chile
| | - Daniel Silva
- Laboratory of Physiology, Hypoxia and Vascular Function, Department of Biomedical Science, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, Chile
| |
Collapse
|
22
|
Norton CE, Segal SS. Calcitonin gene-related peptide hyperpolarizes mouse pulmonary artery endothelial tubes through K ATP channel activation. Am J Physiol Lung Cell Mol Physiol 2018. [PMID: 29543503 DOI: 10.1152/ajplung.00044.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The sensory neurotransmitter calcitonin gene-related peptide (CGRP) is associated with vasodilation of systemic arteries through activation of ATP-sensitive K+ (KATP) channels in smooth muscle cells (SMCs); however, its effects on endothelial cell (EC) membrane potential ( Vm) are unresolved. In pulmonary arteries (PAs) of C57BL/6J mice, we questioned whether CGRP would hyperpolarize ECs as well as SMCs. Intact PAs were isolated and immunostained for CGRP to confirm sensory innervation; vessel segments (1-2 mm long, ∼150 µm diameter) with intact or denuded endothelium were cannulated and pressurized to 16 cmH2O at 37°C. Increasing concentrations (10-10-10-6 M) of CGRP progressively dilated PAs preconstricted with UTP (10-5 M); SMCs hyperpolarized similarly (Δ Vm ∼20 mV) before and after endothelial denudation. To study native intact PA ECs, SMCs were dissociated to isolate endothelial tubes, and their integrity was confirmed by vital dye uptake, nuclear staining, and reproducible electrical and intracellular Ca2+ responses to acetylcholine (10-5 M) over 2 h. Increasing [CGRP] hyperpolarized ECs in a manner similar to SMCs, with each cell layer demonstrating robust immunostaining for CGRP receptor proteins. Increasing concentrations (10-10-10-6 M) of pinacidil, a KATP channel agonist, resulted in progressive hyperpolarization of SMCs of intact PAs (Δ Vm ∼30 mV), which was blocked by glibenclamide (10-6 M), as was hyperpolarization of ECs and SMCs to CGRP. Inhibition of protein kinase A with protein kinase inhibitor (10-5 M) also inhibited hyperpolarization to CGRP. We demonstrate [CGRP]-dependent hyperpolarization of ECs for the first time while validating freshly isolated PA endothelial tubes as an experimental model. Redundant electrical signaling to CGRP in ECs and SMCs implies an integral role for KATP channels in PA dilation.
Collapse
Affiliation(s)
- Charles E Norton
- Department of Medical Pharmacology and Physiology, University of Missouri , Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri , Columbia, Missouri.,Dalton Cardiovascular Research Center , Columbia, Missouri
| |
Collapse
|
23
|
Yadav VR, Song T, Mei L, Joseph L, Zheng YM, Wang YX. PLCγ1-PKCε-IP 3R1 signaling plays an important role in hypoxia-induced calcium response in pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2018; 314:L724-L735. [PMID: 29388468 DOI: 10.1152/ajplung.00243.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxia-induced pulmonary vasoconstriction (HPV) is attributed to an increase in intracellular Ca2+ concentration ([Ca2+]i) in pulmonary artery smooth muscle cells (PASMCs). We have reported that phospholipase C-γ1 (PLCγ1) plays a significant role in the hypoxia-induced increase in [Ca2+]i in PASMCs and attendant HPV. In this study, we intended to determine molecular mechanisms for hypoxic Ca2+ and contractile responses in PASMCs. Our data reveal that hypoxic vasoconstriction occurs in pulmonary arteries, but not in mesenteric arteries. Hypoxia caused a large increase in [Ca2+]i in PASMCs, which is diminished by the PLC inhibitor U73122 and not by its inactive analog U73433 . Hypoxia augments PLCγ1-dependent inositol 1,4,5-trisphosphate (IP3) generation. Exogenous ROS, hydrogen peroxide (H2O2), increases PLCγ1 phosphorylation at tyrosine-783 and IP3 production. IP3 receptor-1 (IP3R1) knock-down remarkably diminishes hypoxia- or H2O2-induced increase in [Ca2+]i. Hypoxia or H2O2 increases the activity of IP3Rs, which is significantly reduced in protein kinase C-ε (PKCε) knockout PASMCs. A higher PLCγ1 expression, activity, and basal [Ca2+]i are found in PASMCs, but not in mesenteric artery smooth muscle cells from mice exposed to chronic hypoxia (CH) for 21 days. CH enhances H2O2- and ATP-induced increase in [Ca2+]i in PASMCs and PLC-dependent, norepinephrine-evoked pulmonary vasoconstriction. In conclusion, acute hypoxia uniquely causes ROS-dependent PLCγ1 activation, IP3 production, PKCε activation, IP3R1 opening, Ca2+ release, and contraction in mouse PASMCs; CH enhances PASM PLCγ1 expression, activity, and function, playing an essential role in pulmonary hypertension in mice.
Collapse
Affiliation(s)
- Vishal R Yadav
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Tengyao Song
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Lin Mei
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Leroy Joseph
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| |
Collapse
|
24
|
Weise-Cross L, Sands MA, Sheak JR, Broughton BRS, Snow JB, Gonzalez Bosc LV, Jernigan NL, Walker BR, Resta TC. Actin polymerization contributes to enhanced pulmonary vasoconstrictor reactivity after chronic hypoxia. Am J Physiol Heart Circ Physiol 2018; 314:H1011-H1021. [PMID: 29373038 DOI: 10.1152/ajpheart.00664.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic hypoxia (CH) augments basal and endothelin-1 (ET-1)-induced pulmonary vasoconstrictor reactivity through reactive oxygen species (ROS) generation and RhoA/Rho kinase (ROCK)-dependent myofilament Ca2+ sensitization. Because ROCK promotes actin polymerization and the actin cytoskeleton regulates smooth muscle tension, we hypothesized that actin polymerization is required for enhanced basal and ET-1-dependent vasoconstriction after CH. To test this hypothesis, both end points were monitored in pressurized, endothelium-disrupted pulmonary arteries (fourth-fifth order) from control and CH (4 wk at 0.5 atm) rats. The actin polymerization inhibitors cytochalasin and latrunculin attenuated both basal and ET-1-induced vasoconstriction only in CH vessels. To test whether CH directly alters the arterial actin profile, we measured filamentous actin (F-actin)-to-globular actin (G-actin) ratios by fluorescent labeling of F-actin and G-actin in fixed pulmonary arteries and actin sedimentation assays using homogenized pulmonary artery lysates. We observed no difference in actin polymerization between groups under baseline conditions, but ET-1 enhanced actin polymerization in pulmonary arteries from CH rats. This response was blunted by the ROS scavenger tiron, the ROCK inhibitor fasudil, and the mDia (RhoA effector) inhibitor small-molecule inhibitor of formin homology domain 2. Immunoblot analysis revealed an effect of CH to increase both phosphorylated (inactive) and total levels of the actin disassembly factor cofilin but not phosphorylated cofilin-to-total cofilin ratios. We conclude that actin polymerization contributes to increased basal pulmonary arterial constriction and ET-1-induced vasoconstrictor reactivity after CH in a ROS- and ROCK-dependent manner. Our results further suggest that enhanced ET-1-mediated actin polymerization after CH is dependent on mDia but independent of changes in the phosphorylated cofilin-to-total cofilin ratio. NEW & NOTEWORTHY This research is the first to demonstrate a role for actin polymerization in chronic hypoxia-induced basal pulmonary arterial constriction and enhanced agonist-induced vasoconstrictor activity. These results suggest that a reactive oxygen species-Rho kinase-actin polymerization signaling pathway mediates this response and may provide a mechanistic basis for the vasoconstrictor component of pulmonary hypertension.
Collapse
Affiliation(s)
- Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Michelle A Sands
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Joshua R Sheak
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Brad R S Broughton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Jessica B Snow
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| |
Collapse
|
25
|
Jiang D, Chen Y, Zhu Y, Fu G, Xu S. Expression of key enzymes in the mevalonate pathway are altered in monocrotaline-induced pulmonary arterial hypertension in rats. Mol Med Rep 2017; 16:9593-9600. [PMID: 29039598 DOI: 10.3892/mmr.2017.7798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 08/31/2017] [Indexed: 11/06/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a serious pulmonary vascular disease. The changes in the structure, function and metabolism of endothelial cells are some of the important features of PAH. Previous studies have demonstrated that the mevalonate pathway is important in cardiovascular remodeling. However, whether the mevalonate pathway is involved in the development of PAH remains to be elucidated. The present study aimed to investigate the expression pattern of mevalonate pathway-related enzymes in monocrotaline (MCT)-induced PAH. F344 rats were randomly divided into two groups (n=6/group): Control group rats were injected with a single dose of saline, and MCT group rats were injected with a single dose of MCT (60 mg/kg). After 4 weeks, the right ventricular systolic pressure (RVSP) was measured, and lung and pulmonary artery tissue samples were collected. It was demonstrated that the RVSP increased and pulmonary vascular remodeling was detected in the PAH group. The expression levels of the enzymes farnesyldiphosphate synthase farnesyltransferase α and geranylgeranyltransferase type I increased in the PAH group, which suggested that the mevalonate pathway may be involved in the pathological development of PAH.
Collapse
Affiliation(s)
- Dongmei Jiang
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Yu Chen
- Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, P.R. China
| | - Yuxiang Zhu
- Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310029, P.R. China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Shiming Xu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
26
|
Cervantes Gracia K, Llanas-Cornejo D, Husi H. CVD and Oxidative Stress. J Clin Med 2017; 6:E22. [PMID: 28230726 PMCID: PMC5332926 DOI: 10.3390/jcm6020022] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/12/2017] [Accepted: 02/14/2017] [Indexed: 12/12/2022] Open
Abstract
Nowadays, it is known that oxidative stress plays at least two roles within the cell, the generation of cellular damage and the involvement in several signaling pathways in its balanced normal state. So far, a substantial amount of time and effort has been expended in the search for a clear link between cardiovascular disease (CVD) and the effects of oxidative stress. Here, we present an overview of the different sources and types of reactive oxygen species in CVD, highlight the relationship between CVD and oxidative stress and discuss the most prominent molecules that play an important role in CVD pathophysiology. Details are given regarding common pharmacological treatments used for cardiovascular distress and how some of them are acting upon ROS-related pathways and molecules. Novel therapies, recently proposed ROS biomarkers, as well as future challenges in the field are addressed. It is apparent that the search for a better understanding of how ROS are contributing to the pathophysiology of CVD is far from over, and new approaches and more suitable biomarkers are needed for the latter to be accomplished.
Collapse
Affiliation(s)
- Karla Cervantes Gracia
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.
| | - Daniel Llanas-Cornejo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.
| | - Holger Husi
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.
| |
Collapse
|
27
|
Jernigan NL, Resta TC, Gonzalez Bosc LV. Altered Redox Balance in the Development of Chronic Hypoxia-induced Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:83-103. [PMID: 29047083 DOI: 10.1007/978-3-319-63245-2_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Normally, the pulmonary circulation is maintained in a low-pressure, low-resistance state with little resting tone. Pulmonary arteries are thin-walled and rely heavily on pulmonary arterial distension and recruitment for reducing pulmonary vascular resistance when cardiac output is elevated. Under pathophysiological conditions, however, active vasoconstriction and vascular remodeling lead to enhanced pulmonary vascular resistance and subsequent pulmonary hypertension (PH). Chronic hypoxia is a critical pathological factor associated with the development of PH resulting from airway obstruction (COPD, sleep apnea), diffusion impairment (interstitial lung disease), developmental lung abnormalities, or high altitude exposure (World Health Organization [WHO]; Group III). The rise in pulmonary vascular resistance increases right heart afterload causing right ventricular hypertrophy that can ultimately lead to right heart failure in patients with chronic lung disease. PH is typically characterized by diminished paracrine release of vasodilators, antimitogenic factors, and antithrombotic factors (e.g., nitric oxide and protacyclin) and enhanced production of vasoconstrictors and mitogenic factors (e.g., reactive oxygen species and endothelin-1) from the endothelium and lung parenchyma. In addition, phenotypic changes to pulmonary arterial smooth muscle cells (PASMC), including alterations in Ca2+ homeostasis, Ca2+ sensitivity, and activation of transcription factors are thought to play prominent roles in the development of both vasoconstrictor and arterial remodeling components of hypoxia-associated PH. These changes in PASMC function are briefly reviewed in Sect. 1 and the influence of altered reactive oxygen species homeostasis on PASMC function discussed in Sects. 2-4.
Collapse
Affiliation(s)
- Nikki L Jernigan
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Thomas C Resta
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Laura V Gonzalez Bosc
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
28
|
Cerny O, Anderson KE, Stephens LR, Hawkins PT, Sebo P. cAMP Signaling of Adenylate Cyclase Toxin Blocks the Oxidative Burst of Neutrophils through Epac-Mediated Inhibition of Phospholipase C Activity. THE JOURNAL OF IMMUNOLOGY 2016; 198:1285-1296. [PMID: 28039302 DOI: 10.4049/jimmunol.1601309] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/16/2016] [Indexed: 01/08/2023]
Abstract
The adenylate cyclase toxin-hemolysin (CyaA) plays a key role in immune evasion and virulence of the whooping cough agent Bordetella pertussis. CyaA penetrates the complement receptor 3-expressing phagocytes and ablates their bactericidal capacities by catalyzing unregulated conversion of cytosolic ATP to the key second messenger molecule cAMP. We show that signaling of CyaA-generated cAMP blocks the oxidative burst capacity of neutrophils by two converging mechanisms. One involves cAMP/protein kinase A-mediated activation of the Src homology region 2 domain-containing phosphatase-1 (SHP-1) and limits the activation of MAPK ERK and p38 that are required for assembly of the NADPH oxidase complex. In parallel, activation of the exchange protein directly activated by cAMP (Epac) provokes inhibition of the phospholipase C by an as yet unknown mechanism. Indeed, selective activation of Epac by the cell-permeable analog 8-(4-chlorophenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate counteracted the direct activation of phospholipase C by 2,4,6-trimethyl-N-[3-(trifluoromethyl)phenyl]benzenesulfonamide. Hence, by inhibiting production of the protein kinase C-activating lipid, diacylglycerol, cAMP/Epac signaling blocks the bottleneck step of the converging pathways of oxidative burst triggering. Manipulation of neutrophil membrane composition by CyaA-produced signaling of cAMP thus enables B. pertussis to evade the key innate host defense mechanism of reactive oxygen species-mediated killing of bacteria by neutrophils.
Collapse
Affiliation(s)
- Ondrej Cerny
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the CAS, v.v.i., 142 20 Prague, Czech Republic; and
| | - Karen E Anderson
- Signalling Programme, Babraham Institute, Babraham, Cambridge CB22 3AT, United Kingdom
| | - Len R Stephens
- Signalling Programme, Babraham Institute, Babraham, Cambridge CB22 3AT, United Kingdom
| | - Phillip T Hawkins
- Signalling Programme, Babraham Institute, Babraham, Cambridge CB22 3AT, United Kingdom
| | - Peter Sebo
- Laboratory of Molecular Biology of Bacterial Pathogens, Institute of Microbiology of the CAS, v.v.i., 142 20 Prague, Czech Republic; and
| |
Collapse
|
29
|
Chen F, Li X, Aquadro E, Haigh S, Zhou J, Stepp DW, Weintraub NL, Barman SA, Fulton DJR. Inhibition of histone deacetylase reduces transcription of NADPH oxidases and ROS production and ameliorates pulmonary arterial hypertension. Free Radic Biol Med 2016; 99:167-178. [PMID: 27498117 PMCID: PMC5240036 DOI: 10.1016/j.freeradbiomed.2016.08.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/30/2016] [Accepted: 08/02/2016] [Indexed: 12/11/2022]
Abstract
Excessive levels of reactive oxygen species (ROS) and increased expression of NADPH oxidases (Nox) have been proposed to contribute to pulmonary artery hypertension (PAH) and other cardiovascular diseases (CVD). Nox enzymes are major sources of ROS but the mechanisms regulating changes in Nox expression in disease states remain poorly understood. Epigenetics encompasses a number of mechanisms that cells employ to regulate the ability to read and transcribe DNA. Histone acetylation is a prominent example of an epigenetic mechanism regulating the expression of numerous genes by altering chromatin accessibility. The goal of this study was to determine whether inhibition of histone deacetylases (HDAC) affects the expression of Nox isoforms and reduces pulmonary hypertension. In immune cells, we found that multiple HDAC inhibitors robustly decreased Nox2 mRNA and protein expression in a dose-dependent manner concomitant with reduced superoxide production. This effect was not restricted to Nox2 as expression of Nox1, Nox4 and Nox5 was also reduced by HDAC inhibition. Surprisingly, Nox promoter-luciferase activity was unchanged in the presence of HDAC inhibitors. In macrophages and lung fibroblasts, ChIP experiments revealed that HDAC inhibitors block the binding of RNA polymerase II and the histone acetyltransferase p300 to the Nox2, Nox4 and Nox5 promoter regions and decrease histones activation marks (H3K4me3 and H3K9ac) at these promoter sites. We further show that the ability of CRISPR-ON to drive transcription of Nox1, Nox2, Nox4 and Nox5 genes is blocked by HDAC inhibitors. In a monocrotaline (MCT) rat model of PAH, multiple HDAC isoforms are upregulated in isolated pulmonary arteries, and HDAC inhibitors attenuate Nox expression in isolated pulmonary arteries and reduce indices of PAH. In conclusion, HDAC inhibitors potently suppress Nox gene expression both in vitro and in vivo via epigenetically regulating chromatin accessibility.
Collapse
Affiliation(s)
- Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029 China; Vascular Biology Center, Augusta University, Augusta, GA 30912, USA.
| | - Xueyi Li
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Emily Aquadro
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Stephen Haigh
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Jiliang Zhou
- Department of Pharmacology, Augusta University, Augusta, GA 30912, USA
| | - David W Stepp
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Neal L Weintraub
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Medicine, Augusta University, Augusta, GA 30912, USA
| | - Scott A Barman
- Department of Pharmacology, Augusta University, Augusta, GA 30912, USA
| | - David J R Fulton
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA; Department of Pharmacology, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
30
|
Rafikova O, Rafikov R, Kangath A, Qu N, Aggarwal S, Sharma S, Desai J, Fields T, Ludewig B, Yuan JXY, Jonigk D, Black SM. Redox regulation of epidermal growth factor receptor signaling during the development of pulmonary hypertension. Free Radic Biol Med 2016; 95:96-111. [PMID: 26928584 PMCID: PMC5929487 DOI: 10.1016/j.freeradbiomed.2016.02.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 11/19/2022]
Abstract
The development of pulmonary hypertension (PH) involves the uncontrolled proliferation of pulmonary smooth muscle cells via increased growth factor receptor signaling. However, the role of epidermal growth factor receptor (EGFR) signaling is controversial, as humans with advanced PH exhibit no changes in EGFR protein levels and purpose of the present study was to determine whether there are post-translational mechanisms that enhance EGFR signaling in PH. The EGFR inhibitor, gefinitib, significantly attenuated EGFR signaling and prevented the development of PH in monocrotaline (MCT)-exposed rats, confirming the contribution of EGFR activation in MCT induced PH. There was an early MCT-mediated increase in hydrogen peroxide, which correlated with the binding of the active metabolite of MCT, monocrotaline pyrrole, to catalase Cys377, disrupting its multimeric structure. This early oxidative stress was responsible for the oxidation of EGFR and the formation of sodium dodecyl sulfate (SDS) stable EGFR dimers through dityrosine cross-linking. These cross-linked dimers exhibited increased EGFR autophosphorylation and signaling. The activation of EGFR signaling did not correlate with pp60(src) dependent Y845 phosphorylation or EGFR ligand expression. Importantly, the analysis of patients with advanced PH revealed the same enhancement of EGFR autophosphorylation and covalent dimer formation in pulmonary arteries, while total EGFR protein levels were unchanged. As in the MCT exposed rat model, the activation of EGFR in human samples was independent of pp60(src) phosphorylation site and ligand expression. This study provides a novel molecular mechanism of oxidative stress stimulated covalent EGFR dimerization via tyrosine dimerization that contributes into development of PH.
Collapse
Affiliation(s)
- Olga Rafikova
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Ruslan Rafikov
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Archana Kangath
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Ning Qu
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Saurabh Aggarwal
- Department of Anesthesiology, University of Alabama, Birmingham, AL, United States
| | - Shruti Sharma
- Center For Biotechnology & Genomic Medicine, Georgia Regents University, Augusta, GA, United States
| | - Julin Desai
- Vascular Biology Center, Georgia Regents University, Augusta, GA, United States
| | - Taylor Fields
- Vascular Biology Center, Georgia Regents University, Augusta, GA, United States
| | - Britta Ludewig
- Institute of Pathology, Hannover Medical School, Hanover, Germany
| | - Jason X-Y Yuan
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hanover, Germany
| | - Stephen M Black
- Department of Medicine, University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
31
|
Wieczfinska J, Sokolowska M, Pawliczak R. NOX Modifiers-Just a Step Away from Application in the Therapy of Airway Inflammation? Antioxid Redox Signal 2015; 23:428-45. [PMID: 24383678 PMCID: PMC4543397 DOI: 10.1089/ars.2013.5783] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE NADPH oxidase (NOX) enzymes, which are widely expressed in different airway cell types, not only contribute to the maintenance of physiological processes in the airways but also participate in the pathogenesis of many acute and chronic diseases. Therefore, the understanding of NOX isoform regulation, expression, and the manner of their potent inhibition might lead to effective therapeutic approaches. RECENT ADVANCES The study of the role of NADPH oxidases family in airway physiology and pathophysiology should be considered as a work in progress. While key questions still remain unresolved, there is significant progress in terms of our understanding of NOX importance in airway diseases as well as a more efficient way of using NOX modifiers in human settings. CRITICAL ISSUES Agents that modify the activity of NADPH enzyme components would be considered useful tools in the treatment of various airway diseases. Nevertheless, profound knowledge of airway pathology, as well as the mechanisms of NOX regulation is needed to develop potent but safe NOX modifiers. FUTURE DIRECTIONS Many compounds seem to be promising candidates for development into useful therapeutic agents, but their clinical potential is yet to be demonstrated. Further analysis of basic mechanisms in human settings, high-throughput compound scanning, clinical trials with new and existing molecules, and the development of new drug delivery approaches are the main directions of future studies on NOX modifiers. In this article, we discuss the current knowledge with regard to NOX isoform expression and regulation in airway inflammatory diseases as well as the aptitudes and therapeutic potential of NOX modifiers.
Collapse
Affiliation(s)
- Joanna Wieczfinska
- 1 Department of Immunopathology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz , Lodz, Poland
| | - Milena Sokolowska
- 2 Critical Care Medicine Department, Clinical Center, National Institutes of Health , Bethesda, Maryland
| | - Rafal Pawliczak
- 1 Department of Immunopathology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz , Lodz, Poland
| |
Collapse
|
32
|
Abstract
PURPOSE OF THE REVIEW The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase with a wide implication in tumor biology, wound healing and development. Besides acting as a growth factor receptor activated by ligands such as EGF, the EGFR can also be transactivated and thereby mediate cross-talk with different signaling pathways. The aim of this review is to illustrate the Janus-faced function of the EGFR in the vasculature with its relevance for vascular biology and disease. RECENT FINDINGS Over recent years, the number of identified signaling partners of the EGFR has steadily increased, as have the biological processes in which the EGFR is thought to be involved. Recently, new models have allowed investigation of EGFR effects in vivo, shedding some light on the overall function of the EGFR in the vasculature. At the same time, EGFR inhibitors and antibodies have become increasingly established in cancer therapy, providing potential therapeutic tools for decreasing EGFR signaling. SUMMARY The EGFR is a versatile signaling pathway integrator associated with vascular homeostasis and disease. In addition to modulating basal vascular tone and tissue homeostasis, the EGFR also seems to be involved in proinflammatory, proliferative, migratory and remodeling processes, with enhanced deposition of extracellular matrix components, thereby promoting vascular diseases such as hypertension or atherosclerosis.
Collapse
|
33
|
Ramiro-Diaz JM, Giermakowska W, Weaver JM, Jernigan NL, Gonzalez Bosc LV. Mechanisms of NFATc3 activation by increased superoxide and reduced hydrogen peroxide in pulmonary arterial smooth muscle. Am J Physiol Cell Physiol 2014; 307:C928-38. [PMID: 25163518 DOI: 10.1152/ajpcell.00244.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We recently demonstrated increased superoxide (O2(·-)) and decreased H2O2 levels in pulmonary arteries of chronic hypoxia-exposed wild-type and normoxic superoxide dismutase 1 (SOD1) knockout mice. We also showed that this reciprocal change in O2(·-) and H2O2 is associated with elevated activity of nuclear factor of activated T cells isoform c3 (NFATc3) in pulmonary arterial smooth muscle cells (PASMC). This suggests that an imbalance in reactive oxygen species levels is required for NFATc3 activation. However, how such imbalance activates NFATc3 is unknown. This study evaluated the importance of O2(·-) and H2O2 in the regulation of NFATc3 activity. We tested the hypothesis that an increase in O2(·-) enhances actin cytoskeleton dynamics and a decrease in H2O2 enhances intracellular Ca(2+) concentration, contributing to NFATc3 nuclear import and activation in PASMC. We demonstrate that, in PASMC, endothelin-1 increases O2(·-) while decreasing H2O2 production through the decrease in SOD1 activity without affecting SOD protein levels. We further demonstrate that O2(·-) promotes, while H2O2 inhibits, NFATc3 activation in PASMC. Additionally, increased O2(·-)-to-H2O2 ratio activates NFATc3, even in the absence of a Gq protein-coupled receptor agonist. Furthermore, O2(·-)-dependent actin polymerization and low intracellular H2O2 concentration-dependent increases in intracellular Ca(2+) concentration contribute to NFATc3 activation. Together, these studies define important and novel regulatory mechanisms of NFATc3 activation in PASMC by reactive oxygen species.
Collapse
Affiliation(s)
- Juan Manuel Ramiro-Diaz
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Wieslawa Giermakowska
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - John M Weaver
- Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico;
| |
Collapse
|
34
|
Chen F, Barman S, Yu Y, Haigh S, Wang Y, Black SM, Rafikov R, Dou H, Bagi Z, Han W, Su Y, Fulton DJR. Caveolin-1 is a negative regulator of NADPH oxidase-derived reactive oxygen species. Free Radic Biol Med 2014; 73:201-13. [PMID: 24835767 PMCID: PMC4228786 DOI: 10.1016/j.freeradbiomed.2014.04.029] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 04/25/2014] [Accepted: 04/27/2014] [Indexed: 01/14/2023]
Abstract
Changes in the expression and function of caveolin-1 (Cav-1) have been proposed as a pathogenic mechanism underlying many cardiovascular diseases. Cav-1 binds to and regulates the activity of numerous signaling proteins via interactions with its scaffolding domain. In endothelial cells, Cav-1 has been shown to reduce reactive oxygen species (ROS) production, but whether Cav-1 regulates the activity of NADPH oxidases (Noxes), a major source of cellular ROS, has not yet been shown. Herein, we show that Cav-1 is primarily expressed in the endothelium and adventitia of pulmonary arteries (PAs) and that Cav-1 expression is reduced in isolated PAs from multiple models of pulmonary artery hypertension (PH). Reduced Cav-1 expression correlates with increased ROS production in the adventitia of hypertensive PA. In vitro experiments revealed a significant ability of Cav-1 and its scaffolding domain to inhibit Nox1-5 activity and it was also found that Cav-1 binds to Nox5 and Nox2 but not Nox4. In addition to posttranslational actions, in primary cells, Cav-1 represses the mRNA and protein expression of Nox2 and Nox4 through inhibition of the NF-κB pathway. Last, in a mouse hypoxia model, the genetic ablation of Cav-1 increased the expression of Nox2 and Nox4 and exacerbated PH. Together, these results suggest that Cav-1 is a negative regulator of Nox function via two distinct mechanisms, acutely through direct binding and chronically through alteration of expression levels. Accordingly, the loss of Cav-1 expression in cardiovascular diseases such as PH may account for the increased Nox activity and greater production of ROS.
Collapse
Affiliation(s)
- Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA.
| | - Scott Barman
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Yanfang Yu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA
| | - Steven Haigh
- Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA
| | - Yusi Wang
- Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA
| | | | | | | | - Zsolt Bagi
- Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA
| | - Weihong Han
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - David J R Fulton
- Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA; Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
35
|
Plomaritas DR, Herbert LM, Yellowhair TR, Resta TC, Gonzalez Bosc LV, Walker BR, Jernigan NL. Chronic hypoxia limits H2O2-induced inhibition of ASIC1-dependent store-operated calcium entry in pulmonary arterial smooth muscle. Am J Physiol Lung Cell Mol Physiol 2014; 307:L419-30. [PMID: 24993130 DOI: 10.1152/ajplung.00095.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
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.
Collapse
Affiliation(s)
- Danielle R Plomaritas
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Lindsay M Herbert
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Tracylyn R Yellowhair
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
36
|
Barman SA, Chen F, Su Y, Dimitropoulou C, Wang Y, Catravas JD, Han W, Orfi L, Szantai-Kis C, Keri G, Szabadkai I, Barabutis N, Rafikova O, Rafikov R, Black SM, Jonigk D, Giannis A, Asmis R, Stepp DW, Ramesh G, Fulton DJR. NADPH oxidase 4 is expressed in pulmonary artery adventitia and contributes to hypertensive vascular remodeling. Arterioscler Thromb Vasc Biol 2014; 34:1704-15. [PMID: 24947524 DOI: 10.1161/atvbaha.114.303848] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Pulmonary hypertension (PH) is a progressive disease arising from remodeling and narrowing of pulmonary arteries (PAs) resulting in high pulmonary blood pressure and ultimately right ventricular failure. Elevated production of reactive oxygen species by NADPH oxidase 4 (Nox4) is associated with increased pressure in PH. However, the cellular location of Nox4 and its contribution to aberrant vascular remodeling in PH remains poorly understood. Therefore, we sought to identify the vascular cells expressing Nox4 in PAs and determine the functional relevance of Nox4 in PH. APPROACH AND RESULTS Elevated expression of Nox4 was detected in hypertensive PAs from 3 rat PH models and human PH using qualititative real-time reverse transcription polymerase chain reaction, Western blot, and immunofluorescence. In the vascular wall, Nox4 was detected in both endothelium and adventitia, and perivascular staining was prominently increased in hypertensive lung sections, colocalizing with cells expressing fibroblast and monocyte markers and matching the adventitial location of reactive oxygen species production. Small-molecule inhibitors of Nox4 reduced adventitial reactive oxygen species generation and vascular remodeling as well as ameliorating right ventricular hypertrophy and noninvasive indices of PA stiffness in monocrotaline-treated rats as determined by morphometric analysis and high-resolution digital ultrasound. Nox4 inhibitors improved PH in both prevention and reversal protocols and reduced the expression of fibroblast markers in isolated PAs. In fibroblasts, Nox4 overexpression stimulated migration and proliferation and was necessary for matrix gene expression. CONCLUSION These findings indicate that Nox4 is prominently expressed in the adventitia and contributes to altered fibroblast behavior, hypertensive vascular remodeling, and development of PH.
Collapse
Affiliation(s)
- Scott A Barman
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary.
| | - Feng Chen
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary.
| | - Yunchao Su
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Christiana Dimitropoulou
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Yusi Wang
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - John D Catravas
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Weihong Han
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Laszlo Orfi
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Csaba Szantai-Kis
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Gyorgy Keri
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Istvan Szabadkai
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Nektarios Barabutis
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Olga Rafikova
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Ruslan Rafikov
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Stephen M Black
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Danny Jonigk
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Athanassios Giannis
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Reto Asmis
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - David W Stepp
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - Ganesan Ramesh
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary
| | - David J R Fulton
- From the Department of Forensic Medicine, Nanjing Medical University, Jiangsu, China (F.C.); Department of Pharmacology and Toxicology (S.A.B., Y.S., W.H., D.J.R.F.) and Vascular Biology Center (F.C., C.D., Y.W., J.D.S., N.B., O.R., R.R., S.M.B., D.W.S., G.R., D.J.R.F.), Georgia Regents University, Augusta; Vichem Chemie, Ltd, Budapest, Hungary (L.O., C.S.-K., G.K., I.S.); Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany (A.G.); Institute for Pathology, Hannover Medical School, Hannover, Germany (D.J.); Departments of Clinical Laboratories and Biochemistry, University of Texas Health Science Center at San Antonio (R.A.); and Pathobiochemical Research Group of Hungarian Academy of Sciences (G.K.) and Department of Pharmaceutical Chemistry (L.O.), Semmelweis University, Budapest, Hungary.
| |
Collapse
|
37
|
Hong Z, Cabrera JA, Mahapatra S, Kutty S, Weir EK, Archer SL. Activation of the EGFR/p38/JNK pathway by mitochondrial-derived hydrogen peroxide contributes to oxygen-induced contraction of ductus arteriosus. J Mol Med (Berl) 2014; 92:995-1007. [PMID: 24906456 DOI: 10.1007/s00109-014-1162-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 03/25/2014] [Accepted: 04/29/2014] [Indexed: 12/17/2022]
Abstract
UNLABELLED Oxygen-induced contraction of the ductus arteriosus (DA) involves a mitochondrial oxygen sensor, which signals pO2 in the DA smooth muscle cell (DASMC) by increasing production of diffusible hydrogen peroxide (H2O2). H2O2 stimulates vasoconstriction by regulating ion channels and Rho kinase, leading to calcium influx and calcium sensitization. Because epidermal growth factor receptor (EGFR) signaling is also redox regulated and participates in oxygen sensing and vasoconstriction in other systems, we explored the role of the EGFR and its signaling cascade (p38 and c-Jun N-amino-terminal kinase (JNK)) in DA contraction. Experiments were performed in DA rings isolated from full-term New Zealand white rabbits and human DASMC. In human DASMCs, increasing pO2 from hypoxia to normoxia (40 to 100 mmHg) significantly increased cytosolic calcium, p < 0.01. This normoxic rise in intracellular calcium was mimicked by EGF and inhibited by EGFR siRNA. In DA rings, EGF caused contraction while the specific EGFR inhibitor (AG1478) and the tyrosine kinase inhibitors (genistein or tyrphostin A23) selectively attenuated oxygen-induced contraction (p < 0.01). Conversely, orthovanadate, a tyrosine phosphatase inhibitor known to activate EGFR signaling, caused dose-dependent contraction of hypoxic DA and superimposed increases in oxygen caused minimal additional contraction. Anisomycin, an activator of EGFR's downstream kinases, p38 and JNK, caused DA contraction; conversely, oxygen-induced DA contraction was blocked by inhibitors of p38 mitogen-activated protein kinases (MAPK) (SB203580) or JNK (JNK inhibitor II). O2-induced phosphorylation of EGFR occurred within 5 min of increasing pO2 and was inhibited by mitochondrial-targeted overexpression of catalase. AG1478 prevented the oxygen-induced p38 and JNK phosphorylation. In conclusion, O2-induced EGFR transactivation initiates p38/JNK-mediated increases in cytosolic calcium and contributes to DA contraction. The EGFR/p38/JNK pathway is regulated by mitochondrial redox signaling and is a promising therapeutic target for modulation of the patent ductus arteriosus. KEY MESSAGES Oxygen activates epidermal growth factor receptor (EGFR) in ductus arteriosus (DA) smooth muscle cells. EGFR inhibition selectively attenuates O2-induced DA constriction. pO2-induced EGFR activation is mediated by mitochondrial-derived hydrogen peroxide. p38 MAPK and JNK mediated EGFR's effects on oxygen-induced DA contraction. Tyrosine kinases and phosphatases participate in oxygen sensing in the DA. The EGFR pathway offers new therapeutic targets to modulate patency of the ductus arteriosus.
Collapse
Affiliation(s)
- Zhigang Hong
- Department of Medicine, Queen's University, Etherington Hall, Room 3041, 94 Stuart St., Kingston, Ontario, K7L 3 N6, Canada
| | | | | | | | | | | |
Collapse
|
38
|
Jernigan NL, Resta TC. Calcium Homeostasis and Sensitization in Pulmonary Arterial Smooth Muscle. Microcirculation 2014; 21:259-71. [DOI: 10.1111/micc.12096] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/25/2013] [Indexed: 01/10/2023]
Affiliation(s)
- Nikki L. Jernigan
- Vascular Physiology Group; Department of Cell Biology and Physiology; University of New Mexico Health Sciences Center; Albuquerque New Mexico USA
| | - Thomas C. Resta
- Vascular Physiology Group; Department of Cell Biology and Physiology; University of New Mexico Health Sciences Center; Albuquerque New Mexico USA
| |
Collapse
|
39
|
Suzuki YJ, Steinhorn RH, Gladwin MT. Antioxidant therapy for the treatment of pulmonary hypertension. Antioxid Redox Signal 2013; 18:1723-6. [PMID: 23330936 PMCID: PMC3941794 DOI: 10.1089/ars.2013.5193] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Substantial experimental evidence suggests the usefulness of antioxidants for the treatment of various forms of pulmonary hypertension. However, no recommendations have yet been made if patients with pulmonary hypertension should receive pharmacologic and/or dietary antioxidants. Our understanding of antioxidants has evolved greatly over the last two decades, from the primitive use of natural antioxidant vitamins to the modulation of vascular oxidases, such as NAD(P)H oxidases. These oxidases and their products not only regulate pulmonary vascular tone and intimal and smooth muscle thickening, but also modulate the adaptation of the right ventricle to increased afterload. It is important that well-designed randomized clinical trials be conducted to test the importance of oxidase-reactive oxygen species activation in the pathogenesis and treatment of pulmonary hypertension. The purpose of this Forum on Pulmonary Hypertension is to summarize the available preclinical information, which may aid in designing and conducting future randomized clinical trials for evaluating the efficacy of antioxidants for the treatment of pulmonary hypertension. The complexity of oxidative pathways contributed to the tremendous difficulties and challenges in selecting agents, doses, and designing clinical trials. Further studies using human, animal, and cell culture models may be needed to define optimal trials. This Forum on Pulmonary Hypertension should stimulate new thinking and provide essential background information to better define the challenges of developing successful randomized clinical trials in the near future.
Collapse
Affiliation(s)
- Yuichiro J. Suzuki
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia
| | - Robin H. Steinhorn
- Department of Pediatrics, University of California at Davis, Sacramento, California
| | - Mark T. Gladwin
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Vascular Medicine Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
40
|
Ramiro-Diaz JM, Nitta CH, Maston LD, Codianni S, Giermakowska W, Resta TC, Gonzalez Bosc LV. NFAT is required for spontaneous pulmonary hypertension in superoxide dismutase 1 knockout mice. Am J Physiol Lung Cell Mol Physiol 2013; 304:L613-25. [PMID: 23475768 PMCID: PMC3652021 DOI: 10.1152/ajplung.00408.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/04/2013] [Indexed: 01/29/2023] Open
Abstract
Elevated reactive oxygen species are implicated in pulmonary hypertension (PH). Superoxide dismutase (SOD) limits superoxide bioavailability, and decreased SOD activity is associated with PH. A decrease in SOD activity is expected to increase superoxide and reduce hydrogen peroxide levels. Such an imbalance of superoxide/hydrogen peroxide has been implicated as a mediator of nuclear factor of activated T cells (NFAT) activation in epidermal cells. We have shown that NFATc3 is required for chronic hypoxia-induced PH. However, it is unknown whether NFATc3 is activated in the pulmonary circulation in a mouse model of decreased SOD1 activity and whether this leads to PH. Therefore, we hypothesized that an elevated pulmonary arterial superoxide/hydrogen peroxide ratio activates NFATc3, leading to PH. We found that SOD1 knockout (KO) mice have elevated pulmonary arterial wall superoxide and decreased hydrogen peroxide levels compared with wild-type (WT) littermates. Right ventricular systolic pressure (RVSP) was elevated in SOD1 KO and was associated with pulmonary arterial remodeling. Vasoreactivity to endothelin-1 was also greater in SOD1 KO vs. WT mice. NFAT activity and NFATc3 nuclear localization were increased in pulmonary arteries from SOD1 KO vs. WT mice. Administration of A-285222 (selective NFAT inhibitor) decreased RVSP, arterial wall thickness, vasoreactivity, and NFAT activity in SOD1 KO mice to WT levels. The SOD mimetic, tempol, also reduced NFAT activity, NFATc3 nuclear localization, and RVSP to WT levels. These findings suggest that an elevated superoxide/hydrogen peroxide ratio activates NFAT in pulmonary arteries, which induces vascular remodeling and increases vascular reactivity leading to PH.
Collapse
Affiliation(s)
- Juan Manuel Ramiro-Diaz
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Strategies Aimed at Nox4 Oxidase Inhibition Employing Peptides from Nox4 B-Loop and C-Terminus and p22 (phox) N-Terminus: An Elusive Target. Int J Hypertens 2013; 2013:842827. [PMID: 23606947 PMCID: PMC3626398 DOI: 10.1155/2013/842827] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/10/2013] [Indexed: 11/22/2022] Open
Abstract
Although NADPH oxidase 4 (Nox4) is the most abundant Nox isoform in systemic vascular endothelial and smooth muscle cells, its function in the vascular tissue is not entirely known. The literature describes a pathophysiological role for Nox4 in cardiovascular disease; however, some studies have reported that it has a protective role. To date, specific Nox4 inhibitors are not available; hence, the development of a pharmacologic tool to assess Nox4's pathophysiological role garners intense interest. In this study, we selected peptides corresponding to regions in the Nox4 oxidase complex critical to holoenzyme activity and postulated their utility as specific competitive inhibitors. Previous studies in our laboratory yielded selective inhibition of Nox2 using this strategy. We postulated that peptides mimicking the Nox4 B-loop and C-terminus and regions on p22phox inhibit Nox4 activity. To test our hypothesis, the inhibitory activity of Nox4 B-loop and C-terminal peptides as well as N-terminal p22phox peptides was assessed in a reconstituted Nox4 system. Our findings demonstrate that Nox4 inhibition is not achieved by preincubation with this comprehensive array of peptides derived from previously identified active regions. These findings suggest that Nox4 exists in a tightly assembled and active conformation which, unlike other Noxes, cannot be disrupted by conventional means.
Collapse
|