1
|
Khan K, Anwar M, Badshah Y, Ashraf NM, Hamid A, Trembley JH, Shabbir M, Afsar T, Husain FM, Khan D, Razak S. Zapotin mitigates breast cancer progression by targeting PKCε mediated glycolytic pathway regulation. BMC Cancer 2025; 25:798. [PMID: 40296014 PMCID: PMC12039207 DOI: 10.1186/s12885-025-14202-z] [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: 01/28/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The breast cancer recurrence and chemoresistance has increased over the years. A novel PKC, PKCε, may promote chemoresistance by causing hypoxia and cancer metabolic rewiring. A natural flavonoid, Zapotin, in colon cancer cells may modulate PKCε expression. Therefore, this study aimed to explore Zapotin impact on PKCε expression and the metabolic profile of breast cancer cells. METHODS Pharmacophore analysis of Zapotin was performed and molecular dynamics (MD) simulations were employed to study PKCε and Zapotin interaction stability. The effect of Zapotin treatment on PKCε expression and various aspects of cancer cell viability and metabolism was studied in MCF-7 and MDA-MB-231 breast cancer cell lines using real-time PCR, growth and death assays, and Gas Chromatography-Mass Spectrometry. RESULTS In silico analyses revealed good solubility and absorption of Zapotin with lower toxicity. Zapotin showed cancer cell-specific cytotoxicity (P < 0.0001). It's treatment also reduced breast cancer cell viability, colony formation, and migratory potential by targeting PKCε and associated HIF-1ɑ and VEGF signaling (P < 0.01). Zapotin also impacted PKCε-mediated metabolic signaling by targeting glycolytic pathways. CONCLUSION This study demonstrated the role of PKCε mediated HIF-1ɑ, VEGF, and glycolytic pathways in promoting breast carcinogenicity and demonstrated Zapotin as a potential treatment option for different types of breast tumors.
Collapse
Affiliation(s)
- Khushbukhat Khan
- Department of Biomedicine, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Maryam Anwar
- Department of Biomedicine, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Yasmin Badshah
- Department of Biomedicine, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Naeem Mahmood Ashraf
- School of Biochemistry & Biotechnology, University of the Punjab, Lahore, Pakistan.
| | - Arslan Hamid
- LIMES Institute (AG-Netea), University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Janeen H Trembley
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Minneapolis VA Health Care System Research Service, Minneapolis, MN, USA
| | - Maria Shabbir
- Department of Biomedicine, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Tayyaba Afsar
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Dilawar Khan
- Department of Biomedicine, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, 44000, Pakistan
| | - Suhail Razak
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
2
|
Zhang J, Yan H, Wang Y, Yue X, Wang M, Liu L, Qiao P, Zhu Y, Li Z. Emerging insights into pulmonary hypertension: the potential role of mitochondrial dysfunction and redox homeostasis. Mol Cell Biochem 2025; 480:1407-1429. [PMID: 39254871 DOI: 10.1007/s11010-024-05096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024]
Abstract
Pulmonary hypertension (PH) is heterogeneous diseases that can lead to death due to progressive right heart failure. Emerging evidence suggests that, in addition to its role in ATP production, changes in mitochondrial play a central role in their pathogenesis, regulating integrated metabolic and signal transduction pathways. This review focuses on the basic principles of mitochondrial redox status in pulmonary vascular and right ventricular disorders, a series of dysfunctional processes including mitochondrial quality control (mitochondrial biogenesis, mitophagy, mitochondrial dynamics, mitochondrial unfolded protein response) and mitochondrial redox homeostasis. In addition, we will summarize how mitochondrial renewal and dynamic changes provide innovative insights for studying and evaluating PH. This will provide us with a clearer understanding of the initial signal transmission of mitochondria in PH, which would further improve our understanding of the pathogenesis of PH.
Collapse
Affiliation(s)
- Junming Zhang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Huimin Yan
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Yan Wang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Xian Yue
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Meng Wang
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Limin Liu
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Pengfei Qiao
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Yixuan Zhu
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China
| | - Zhichao Li
- Faculty of Life Science & Medicine, Northwest University, Xi'an, 710127, Shaanxi, China.
| |
Collapse
|
3
|
Wang H, Song TY, Reyes-García J, Wang YX. Hypoxia-Induced Mitochondrial ROS and Function in Pulmonary Arterial Endothelial Cells. Cells 2024; 13:1807. [PMID: 39513914 PMCID: PMC11545379 DOI: 10.3390/cells13211807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Pulmonary artery endothelial cells (PAECs) are a major contributor to hypoxic pulmonary hypertension (PH) due to the possible roles of reactive oxygen species (ROS). However, the molecular mechanisms and functional roles of ROS in PAECs are not well established. In this study, we first used Amplex UltraRed reagent to assess hydrogen peroxide (H2O2) generation. The result indicated that hypoxic exposure resulted in a significant increase in Amplex UltraRed-derived fluorescence (i.e., H2O2 production) in human PAECs. To complement this result, we employed lucigenin as a probe to detect superoxide (O2-) production. Our assays showed that hypoxia largely increased O2- production. Hypoxia also enhanced H2O2 production in the mitochondria from PAECs. Using the genetically encoded H2O2 sensor HyPer, we further revealed the hypoxic ROS production in PAECs, which was fully blocked by the mitochondrial inhibitor rotenone or myxothiazol. Interestingly, hypoxia caused an increase in the migration of PAECs, determined by scratch wound assay. In contrast, nicotine, a major cigarette or e-cigarette component, had no effect. Moreover, hypoxia and nicotine co-exposure further increased migration. Transfection of lentiviral shRNAs specific for the mitochondrial Rieske iron-sulfur protein (RISP), which knocked down its expression and associated ROS generation, inhibited the hypoxic migration of PAECs. Hypoxia largely increased the proliferation of PAECs, determined using Ki67 staining and direct cell number accounting. Similarly, nicotine caused a large increase in proliferation. Moreover, hypoxia/nicotine co-exposure elicited a further increase in cell proliferation. RISP knockdown inhibited the proliferation of PAECs following hypoxia, nicotine exposure, and hypoxia/nicotine co-exposure. Taken together, our data demonstrate that hypoxia increases RISP-mediated mitochondrial ROS production, migration, and proliferation in human PAECs; nicotine has no effect on migration, increases proliferation, and promotes hypoxic proliferation; the effects of nicotine are largely mediated by RISP-dependent mitochondrial ROS signaling. Conceivably, PAECs may contribute to PH via the RISP-mediated mitochondrial ROS.
Collapse
Affiliation(s)
- Harrison Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY 12208, USA (T.-Y.S.); (J.R.-G.)
| | - Teng-Yao Song
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY 12208, USA (T.-Y.S.); (J.R.-G.)
| | - Jorge Reyes-García
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY 12208, USA (T.-Y.S.); (J.R.-G.)
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY 12208, USA (T.-Y.S.); (J.R.-G.)
| |
Collapse
|
4
|
Geng Y, Hu Y, Zhang F, Tuo Y, Ge R, Bai Z. Mitochondria in hypoxic pulmonary hypertension, roles and the potential targets. Front Physiol 2023; 14:1239643. [PMID: 37645564 PMCID: PMC10461481 DOI: 10.3389/fphys.2023.1239643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023] Open
Abstract
Mitochondria are the centrol hub for cellular energy metabolisms. They regulate fuel metabolism by oxygen levels, participate in physiological signaling pathways, and act as oxygen sensors. Once oxygen deprived, the fuel utilizations can be switched from mitochondrial oxidative phosphorylation to glycolysis for ATP production. Notably, mitochondria can also adapt to hypoxia by making various functional and phenotypes changes to meet the demanding of oxygen levels. Hypoxic pulmonary hypertension is a life-threatening disease, but its exact pathgenesis mechanism is still unclear and there is no effective treatment available until now. Ample of evidence indicated that mitochondria play key factor in the development of hypoxic pulmonary hypertension. By hypoxia-inducible factors, multiple cells sense and transmit hypoxia signals, which then control the expression of various metabolic genes. This activation of hypoxia-inducible factors considered associations with crosstalk between hypoxia and altered mitochondrial metabolism, which plays an important role in the development of hypoxic pulmonary hypertension. Here, we review the molecular mechanisms of how hypoxia affects mitochondrial function, including mitochondrial biosynthesis, reactive oxygen homeostasis, and mitochondrial dynamics, to explore the potential of improving mitochondrial function as a strategy for treating hypoxic pulmonary hypertension.
Collapse
Affiliation(s)
- Yumei Geng
- Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Research Center for High Altitude Medicine, Qinghai University, Xining, China
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People’s Hospital, Xining, China
| | - Yu Hu
- Department of Pharmacy, Qinghai Provincial Traffic Hospital, Xining, China
| | - Fang Zhang
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People’s Hospital, Xining, China
| | - Yajun Tuo
- Department of Respiratory and Critical Care Medicine, Qinghai Provincial People’s Hospital, Xining, China
| | - Rili Ge
- Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Research Center for High Altitude Medicine, Qinghai University, Xining, China
| | - Zhenzhong Bai
- Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Research Center for High Altitude Medicine, Qinghai University, Xining, China
| |
Collapse
|
5
|
Wang YX, Reyes-García J, Di Mise A, Zheng YM. Role of ryanodine receptor 2 and FK506-binding protein 12.6 dissociation in pulmonary hypertension. J Gen Physiol 2023; 155:e202213100. [PMID: 36625865 PMCID: PMC9836826 DOI: 10.1085/jgp.202213100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 07/29/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by a progressive increase in pulmonary arterial pressure leading to right ventricular failure and death. A major cellular response in this disease is the contraction of smooth muscle cells (SMCs) of the pulmonary vasculature. Cell contraction is determined by the increase in intracellular Ca2+ concentration ([Ca2+]i), which is generated and regulated by various ion channels. Several studies by us and others have shown that ryanodine receptor 2 (RyR2), a Ca2+-releasing channel in the sarcoplasmic reticulum (SR), is an essential ion channel for the control of [Ca2+]i in pulmonary artery SMCs (PASMCs), thereby mediating the sustained vasoconstriction seen in PH. FK506-binding protein 12.6 (FKBP12.6) strongly associates with RyR2 to stabilize its functional activity. FKBP12.6 can be dissociated from RyR2 by a hypoxic stimulus to increase channel function and Ca2+ release, leading to pulmonary vasoconstriction and PH. More specifically, dissociation of the RyR2-FKBP12.6 complex is a consequence of increased mitochondrial ROS generation mediated by the Rieske iron-sulfur protein (RISP) at the mitochondrial complex III after hypoxia. Overall, RyR2/FKBP12.6 dissociation and the corresponding signaling pathway may be an important factor in the development of PH. Novel drugs and biologics targeting RyR2, FKBP12.6, and related molecules may become unique effective therapeutics for PH.
Collapse
Affiliation(s)
- Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Jorge Reyes-García
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México,Ciudad de México, México
| | - Annarita Di Mise
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
6
|
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
|
7
|
Jain PP, Hosokawa S, Babicheva A, Zhao T, Chen J, Thistlethwaite PA, Makino A, Yuan JXJ. In Vivo and Ex Vivo Experimental Approach for Studying Functional Role of Notch in Pulmonary Vascular Disease. Methods Mol Biol 2022; 2472:209-220. [PMID: 35674903 DOI: 10.1007/978-1-0716-2201-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease characterized by sustained vasoconstriction, concentric wall thickening and vascular remodeling leading to increased pulmonary vascular resistance, causing right heart failure and death. Acute alveolar hypoxia causes pulmonary vasoconstriction, while sustained hypoxia causes pulmonary hypertension (PH). Activation of Notch signaling is implicated in the development of PAH and chronic hypoxia induced PH via partially its enhancing effect on Ca2+ signaling in pulmonary arterial smooth muscle cells (PASMCs). Pharmacological experiments and genetic approach using animal models of experimental PH (e.g., chronic hypoxia-induced PH) have been routinely utilized to study pathogenic mechanisms of PAH/PH and identify novel therapeutic targets. In this chapter, we describe protocols to investigate the role of Notch by measuring pulmonary hemodynamics in vivo and pulmonary arterial pressure ex vivo in mouse models of experimental PH. Using these experimental protocols, one can study the role of Notch or Notch signaling pathway in the pathogenic mechanisms of pulmonary vascular disease and develop novel therapies by targeting Notch ligands and receptors.
Collapse
Affiliation(s)
- Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Susumu Hosokawa
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Multi-Omics Approach Profiling Metabolic Remodeling in Early Systolic Dysfunction and in Overt Systolic Heart Failure. Int J Mol Sci 2021; 23:ijms23010235. [PMID: 35008662 PMCID: PMC8745344 DOI: 10.3390/ijms23010235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/06/2021] [Accepted: 12/22/2021] [Indexed: 01/19/2023] Open
Abstract
Metabolic remodeling plays an important role in the pathophysiology of heart failure (HF). We sought to characterize metabolic remodeling and implicated signaling pathways in two rat models of early systolic dysfunction (MOD), and overt systolic HF (SHF). Tandem mass tag-labeled shotgun proteomics, phospho-(p)-proteomics, and non-targeted metabolomics analyses were performed in left ventricular myocardium tissue from Sham, MOD, and SHF using liquid chromatography–mass spectrometry, n = 3 biological samples per group. Mitochondrial proteins were predominantly down-regulated in MOD (125) and SHF (328) vs. Sham. Of these, 82% (103/125) and 66% (218/328) were involved in metabolism and respiration. Oxidative phosphorylation, mitochondrial fatty acid β-oxidation, Krebs cycle, branched-chain amino acids, and amino acid (glutamine and tryptophan) degradation were highly enriched metabolic pathways that decreased in SHF > MOD. Glycogen and glucose degradation increased predominantly in MOD, whereas glycolysis and pyruvate metabolism decreased predominantly in SHF. PKA signaling at the endoplasmic reticulum–mt interface was attenuated in MOD, whereas overall PKA and AMPK cellular signaling were attenuated in SHF vs. Sham. In conclusion, metabolic remodeling plays an important role in myocardial remodeling. PKA and AMPK signaling crosstalk governs metabolic remodeling in progression to SHF.
Collapse
|
9
|
Wang M, Liu Y, Liang Y, Naruse K, Takahashi K. Systematic Understanding of Pathophysiological Mechanisms of Oxidative Stress-Related Conditions-Diabetes Mellitus, Cardiovascular Diseases, and Ischemia-Reperfusion Injury. Front Cardiovasc Med 2021; 8:649785. [PMID: 33928135 PMCID: PMC8076504 DOI: 10.3389/fcvm.2021.649785] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) plays a role in intracellular signal transduction under physiological conditions while also playing an essential role in diseases such as hypertension, ischemic heart disease, and diabetes, as well as in the process of aging. The influence of ROS has some influence on the frequent occurrence of cardiovascular diseases (CVD) in diabetic patients. In this review, we considered the pathophysiological relationship between diabetes and CVD from the perspective of ROS. In addition, considering organ damage due to ROS elevation during ischemia-reperfusion, we discussed heart and lung injuries. Furthermore, we have focused on the transient receptor potential (TRP) channels and L-type calcium channels as molecular targets for ROS in ROS-induced tissue damages and have discussed about the pathophysiological mechanism of the injury.
Collapse
Affiliation(s)
| | | | | | | | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
10
|
Li XQ, Zheng YM, Reyes-García J, Wang YX. Diversity of ryanodine receptor 1-mediated Ca 2+ signaling in systemic and pulmonary artery smooth muscle cells. Life Sci 2021; 270:119016. [PMID: 33515564 DOI: 10.1016/j.lfs.2021.119016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 12/26/2020] [Accepted: 01/03/2021] [Indexed: 11/26/2022]
Abstract
AIMS Ryanodine receptor-1 (RyR1) is essential for skeletal muscle cell functions. However, its roles in vascular smooth muscle cells (SMCs) are well recognized. This study aims to determine the potential physiological importance and difference in systemic and pulmonary artery SMCs (SASMCs and PASMCs). METHODS Local and global Ca2+ release were measured using a laser scanning confocal microscope and wide-field fluorescence microscope; membrane currents were recorded using a patch clamp recording; muscle contraction was determined using an organ bath system; RyR protein expression was assessed using immunofluorescence staining. Homozygous and heterozygous RyR1 gene knockout (RyR1-/- and RyR1+/-) mice were used to determine its specific functions. KEY FINDINGS Ca2+ sparks were more prominently decreased in RyR1-/- ASMCs than in PASMCs. Caffeine induced a smaller increase in [Ca2+]i in both RyR1+/+ and RyR1-/- ASMCs than in PASMCs. High K+ produced a reduced [Ca2+]i increase in RyR1-/- PASMCs and ASMCs as well as a reduced contraction in RyR1+/- pulmonary artery and aortic tissues. ATP elicited a smaller increase in [Ca2+]i in RyR1-/- ASMCs and PASMCs with a greater inhibition in ASMCs. Norepinephrine-elicited muscle contraction was reduced in RyR1+/- aortic and pulmonary arteries. IP3 dialysis-induced Ca2+ release was much smaller in RyR1+/- ASMCs and PASMCs. Hypoxia-induced large Ca2+ and contractile responses were inhibited in RyR1+/- PASMCs. However, hypoxic exposure did not evoke a notable increase in [Ca2+]i in ASMCs. SIGNIFICANCE Our findings for the first time provide clear genetic evidence for the functional importance and difference of RyR1 in systemic and pulmonary artery SMCs.
Collapse
Affiliation(s)
- Xiao-Qiang Li
- Albany Medical College, Department of Molecular & Cellular Physiology (MC-8), 47 New Scotland Avenue, Albany, NY 12208, United States of America
| | - Yun-Min Zheng
- Albany Medical College, Department of Molecular & Cellular Physiology (MC-8), 47 New Scotland Avenue, Albany, NY 12208, United States of America
| | - Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, México
| | - Yong-Xiao Wang
- Albany Medical College, Department of Molecular & Cellular Physiology (MC-8), 47 New Scotland Avenue, Albany, NY 12208, United States of America.
| |
Collapse
|
11
|
Kotrasová V, Keresztesová B, Ondrovičová G, Bauer JA, Havalová H, Pevala V, Kutejová E, Kunová N. Mitochondrial Kinases and the Role of Mitochondrial Protein Phosphorylation in Health and Disease. Life (Basel) 2021; 11:life11020082. [PMID: 33498615 PMCID: PMC7912454 DOI: 10.3390/life11020082] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
The major role of mitochondria is to provide cells with energy, but no less important are their roles in responding to various stress factors and the metabolic changes and pathological processes that might occur inside and outside the cells. The post-translational modification of proteins is a fast and efficient way for cells to adapt to ever changing conditions. Phosphorylation is a post-translational modification that signals these changes and propagates these signals throughout the whole cell, but it also changes the structure, function and interaction of individual proteins. In this review, we summarize the influence of kinases, the proteins responsible for phosphorylation, on mitochondrial biogenesis under various cellular conditions. We focus on their role in keeping mitochondria fully functional in healthy cells and also on the changes in mitochondrial structure and function that occur in pathological processes arising from the phosphorylation of mitochondrial proteins.
Collapse
Affiliation(s)
- Veronika Kotrasová
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská Cesta 21, 845 51 Bratislava, Slovakia; (V.K.); (B.K.); (G.O.); (J.A.B.); (H.H.); (V.P.)
| | - Barbora Keresztesová
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská Cesta 21, 845 51 Bratislava, Slovakia; (V.K.); (B.K.); (G.O.); (J.A.B.); (H.H.); (V.P.)
- First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, 128 00 Prague, Czech Republic
| | - Gabriela Ondrovičová
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská Cesta 21, 845 51 Bratislava, Slovakia; (V.K.); (B.K.); (G.O.); (J.A.B.); (H.H.); (V.P.)
| | - Jacob A. Bauer
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská Cesta 21, 845 51 Bratislava, Slovakia; (V.K.); (B.K.); (G.O.); (J.A.B.); (H.H.); (V.P.)
| | - Henrieta Havalová
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská Cesta 21, 845 51 Bratislava, Slovakia; (V.K.); (B.K.); (G.O.); (J.A.B.); (H.H.); (V.P.)
| | - Vladimír Pevala
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská Cesta 21, 845 51 Bratislava, Slovakia; (V.K.); (B.K.); (G.O.); (J.A.B.); (H.H.); (V.P.)
| | - Eva Kutejová
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská Cesta 21, 845 51 Bratislava, Slovakia; (V.K.); (B.K.); (G.O.); (J.A.B.); (H.H.); (V.P.)
- Correspondence: (E.K.); (N.K.)
| | - Nina Kunová
- Institute of Molecular Biology, Slovak Academy of Sciences, Dúbravská Cesta 21, 845 51 Bratislava, Slovakia; (V.K.); (B.K.); (G.O.); (J.A.B.); (H.H.); (V.P.)
- First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, 128 00 Prague, Czech Republic
- Correspondence: (E.K.); (N.K.)
| |
Collapse
|
12
|
Maietta V, Reyes-García J, Yadav VR, Zheng YM, Peng X, Wang YX. Cellular and Molecular Processes in Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:21-38. [PMID: 34019261 DOI: 10.1007/978-3-030-68748-9_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pulmonary hypertension (PH) is a progressive lung disease characterized by persistent pulmonary vasoconstriction. Another well-recognized characteristic of PH is the muscularization of peripheral pulmonary arteries. This pulmonary vasoremodeling manifests in medial hypertrophy/hyperplasia of smooth muscle cells (SMCs) with possible neointimal formation. The underlying molecular processes for these two major vascular responses remain not fully understood. On the other hand, a series of very recent studies have shown that the increased reactive oxygen species (ROS) seems to be an important player in mediating pulmonary vasoconstriction and vasoremodeling, thereby leading to PH. Mitochondria are a primary site for ROS production in pulmonary artery (PA) SMCs, which subsequently activate NADPH oxidase to induce further ROS generation, i.e., ROS-induced ROS generation. ROS control the activity of multiple ion channels to induce intracellular Ca2+ release and extracellular Ca2+ influx (ROS-induced Ca2+ release and influx) to cause PH. ROS and Ca2+ signaling may synergistically trigger an inflammatory cascade to implicate in PH. Accordingly, this paper explores the important roles of ROS, Ca2+, and inflammatory signaling in the development of PH, including their reciprocal interactions, key molecules, and possible therapeutic targets.
Collapse
Affiliation(s)
- Vic Maietta
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Jorge Reyes-García
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA.,Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Vishal R Yadav
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Yun-Min Zheng
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA.
| | - Xu Peng
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, TX, USA.
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
13
|
Jain PP, Hosokawa S, Xiong M, Babicheva A, Zhao T, Rodriguez M, Rahimi S, Pourhashemi K, Balistrieri F, Lai N, Malhotra A, Shyy JYJ, Valdez-Jasso D, Thistlethwaite PA, Makino A, Yuan JXJ. Revisiting the mechanism of hypoxic pulmonary vasoconstriction using isolated perfused/ventilated mouse lung. Pulm Circ 2020; 10:2045894020956592. [PMID: 33282184 PMCID: PMC7691930 DOI: 10.1177/2045894020956592] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/16/2020] [Indexed: 12/13/2022] Open
Abstract
Hypoxic Pulmonary Vasoconstriction (HPV) is an important physiological mechanism of the lungs that matches perfusion to ventilation thus maximizing O2 saturation of the venous blood within the lungs. This study emphasizes on principal pathways in the initiation and modulation of hypoxic pulmonary vasoconstriction with a primary focus on the role of Ca2+ signaling and Ca2+ influx pathways in hypoxic pulmonary vasoconstriction. We used an ex vivo model, isolated perfused/ventilated mouse lung to evaluate hypoxic pulmonary vasoconstriction. Alveolar hypoxia (utilizing a mini ventilator) rapidly and reversibly increased pulmonary arterial pressure due to hypoxic pulmonary vasoconstriction in the isolated perfused/ventilated lung. By applying specific inhibitors for different membrane receptors and ion channels through intrapulmonary perfusion solution in isolated lung, we were able to define the targeted receptors and channels that regulate hypoxic pulmonary vasoconstriction. We show that extracellular Ca2+ or Ca2+ influx through various Ca2+-permeable channels in the plasma membrane is required for hypoxic pulmonary vasoconstriction. Removal of extracellular Ca2+ abolished hypoxic pulmonary vasoconstriction, while blockade of L-type voltage-dependent Ca2+ channels (with nifedipine), non-selective cation channels (with 30 µM SKF-96365), and TRPC6/TRPV1 channels (with 1 µM SAR-7334 and 30 µM capsazepine, respectively) significantly and reversibly inhibited hypoxic pulmonary vasoconstriction. Furthermore, blockers of Ca2+-sensing receptors (by 30 µM NPS2143, an allosteric Ca2+-sensing receptors inhibitor) and Notch (by 30 µM DAPT, a γ-secretase inhibitor) also attenuated hypoxic pulmonary vasoconstriction. These data indicate that Ca2+ influx in pulmonary arterial smooth muscle cells through voltage-dependent, receptor-operated, and store-operated Ca2+ entry pathways all contribute to initiation of hypoxic pulmonary vasoconstriction. The extracellular Ca2+-mediated activation of Ca2+-sensing receptors and the cell-cell interaction via Notch ligands and receptors contribute to the regulation of hypoxic pulmonary vasoconstriction.
Collapse
Affiliation(s)
- Pritesh P. Jain
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Susumu Hosokawa
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
- Department of Pediatrics, Tokyo Medical
and Dental University, Tokyo, Japan
| | - Mingmei Xiong
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
- Department of Critical Medicine, The
Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Tengteng Zhao
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Marisela Rodriguez
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Shamin Rahimi
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Kiana Pourhashemi
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Francesca Balistrieri
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Ning Lai
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Atul Malhotra
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - John Y.-J. Shyy
- Division of Cardiovascular Medicine,
Department of Medicine, University of California, San Diego, USA
| | | | | | - Ayako Makino
- Division of Endocrinology and
Metabolism, University of California, San Diego, CA, USA
| | - Jason X.-J. Yuan
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| |
Collapse
|
14
|
Yang Z, Song T, Truong L, Reyes-García J, Wang L, Zheng YM, Wang YX. Important Role of Sarcoplasmic Reticulum Ca 2+ Release via Ryanodine Receptor-2 Channel in Hypoxia-Induced Rieske Iron-Sulfur Protein-Mediated Mitochondrial Reactive Oxygen Species Generation in Pulmonary Artery Smooth Muscle Cells. Antioxid Redox Signal 2020; 32:447-462. [PMID: 31456413 PMCID: PMC6987675 DOI: 10.1089/ars.2018.7652] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Aims: It is known that mitochondrial reactive oxygen species generation ([ROS]m) causes the release of Ca2+via ryanodine receptor-2 (RyR2) on the sarcoplasmic reticulum (SR) in pulmonary artery smooth muscle cells (PASMCs), playing an essential role in hypoxic pulmonary vasoconstriction (HPV). In this study, we sought to determine whether hypoxia-induced RyR2-mediated Ca2+ release may in turn promote [ROS]m in PASMCs and the underlying signaling mechanism. Results: Our data reveal that application of caffeine or norepinephrine to induce Ca2+ release increased [ROS]m in PASMCs. Likewise, exogenous Ca2+ augmented ROS generation in isolated mitochondria and at complex III from PASMCs. Inhibition of mitochondrial Ca2+ uniporter (MCU) with Ru360 attenuated agonist-induced [ROS]m. Ru360 produced a similar inhibitory effect on hypoxia-induced [ROS]m. Rieske iron-sulfur protein (RISP) gene knockdown inhibited Ca2+- and caffeine-induced [ROS]m. Inhibition of RyR2 by tetracaine or RyR2 gene knockout suppressed hypoxia-induced [ROS]m as well. Innovation: In this article, we present convincing evidence that Ca2+ release following hypoxia or RyR simulation causes a significant increase in MCU, and the increased MCU subsequently RISP-dependent [ROS]m, which provides a positive feedback mechanism to enhance hypoxia-initiated [ROS]m in PASMCs. Conclusion: Our findings demonstrate that hypoxia-induced mitochondrial ROS-dependent SR RyR2-mediated Ca2+ release increases MCU and then RISP-dependent [ROS]m in PASMCs, which may make significant contributions to HPV and associated pulmonary hypertension.
Collapse
Affiliation(s)
- Zhao Yang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York.,Department of Respiratory Medicine, Suzhou Science & Technology Town Hospital, Suzhou, China
| | - Tengyao Song
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Lillian Truong
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Jorge Reyes-García
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Lan Wang
- 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
|
15
|
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: 3.0] [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
|
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: 71] [Impact Index Per Article: 11.8] [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
|
Lucero M, Suarez AE, Chambers JW. Phosphoregulation on mitochondria: Integration of cell and organelle responses. CNS Neurosci Ther 2019; 25:837-858. [PMID: 31025544 PMCID: PMC6566066 DOI: 10.1111/cns.13141] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are highly integrated organelles that are crucial to cell adaptation and mitigating adverse physiology. Recent studies demonstrate that fundamental signal transduction pathways incorporate mitochondrial substrates into their biological programs. Reversible phosphorylation is emerging as a useful mechanism to modulate mitochondrial function in accordance with cellular changes. Critical serine/threonine protein kinases, such as the c-Jun N-terminal kinase (JNK), protein kinase A (PKA), PTEN-induced kinase-1 (PINK1), and AMP-dependent protein kinase (AMPK), readily translocate to the outer mitochondrial membrane (OMM), the interface of mitochondria-cell communication. OMM protein kinases phosphorylate diverse mitochondrial substrates that have discrete effects on organelle dynamics, protein import, respiratory complex activity, antioxidant capacity, and apoptosis. OMM phosphorylation events can be tempered through the actions of local protein phosphatases, such as mitogen-activated protein kinase phosphatase-1 (MKP-1) and protein phosphatase 2A (PP2A), to regulate the extent and duration of signaling. The central mediators of OMM signal transduction are the scaffold proteins because the relative abundance of these accessory proteins determines the magnitude and duration of a signaling event on the mitochondrial surface, which dictates the biological outcome of a local signal transduction pathway. The concentrations of scaffold proteins, such as A-kinase anchoring proteins (AKAPs) and Sab (or SH3 binding protein 5-SH3BP5), have been shown to influence neuronal survival and vulnerability, respectively, in models of Parkinson's disease (PD), highlighting the importance of OMM signaling to health and disease. Despite recent progress, much remains to be discovered concerning the mechanisms of OMM signaling. Nonetheless, enhancing beneficial OMM signaling events and inhibiting detrimental protein-protein interactions on the mitochondrial surface may represent highly selective approaches to restore mitochondrial health and homeostasis and mitigate organelle dysfunction in conditions such as PD.
Collapse
Affiliation(s)
- Maribel Lucero
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, the Biomolecular Sciences Institute, Florida International University, Miami, Florida
| | - Ana E Suarez
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, the Biomolecular Sciences Institute, Florida International University, Miami, Florida
| | - Jeremy W Chambers
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, the Biomolecular Sciences Institute, Florida International University, Miami, Florida
| |
Collapse
|
18
|
Antimycin A-induced mitochondrial dysfunction activates vagal sensory neurons via ROS-dependent activation of TRPA1 and ROS-independent activation of TRPV1. Brain Res 2019; 1715:94-105. [PMID: 30914247 DOI: 10.1016/j.brainres.2019.03.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/11/2019] [Accepted: 03/23/2019] [Indexed: 01/10/2023]
Abstract
Inflammation causes activation of nociceptive sensory nerves, resulting in debilitating sensations and reflexes. Inflammation also induces mitochondrial dysfunction through multiple mechanisms. Sensory nerve terminals are densely packed with mitochondria, suggesting that mitochondrial signaling may play a role in inflammation-induced nociception. We have previously shown that agents that induce mitochondrial dysfunction, such as antimycin A, activate a subset of nociceptive vagal sensory nerves that express transient receptor potential (TRP) channels ankyrin 1 (A1) and vanilloid 1 (V1). However, the mechanisms underlying these responses are incompletely understood. Here, we studied the contribution of TRPA1, TRPV1 and reactive oxygen species (ROS) to antimycin A-induced vagal sensory nerve activation in dissociated neurons and at the sensory terminals of bronchopulmonary C-fibers. Nociceptive neurons were defined chemically and genetically. Antimycin A-evoked activation of vagal nociceptors in a Fura2 Ca2+ assay correlated with TRPV1 responses compared to TRPA1 responses. Nociceptor activation was dependent on both TRP channels, with TRPV1 predominating in a majority of responding nociceptors and TRPA1 predominating only in nociceptors with the greatest responses. Surprisingly, both TRPA1 and TRPV1 were activated by H2O2 when expressed in HEK293. Nevertheless, targeting ROS had no effect of antimycin A-evoked TRPV1 activation in either HEK293 or vagal neurons. In contrast, targeting ROS inhibited antimycin A-evoked TRPA1 activation in HEK293, vagal neurons and bronchopulmonary C-fibers, and a ROS-insensitive TRPA1 mutant was completely insensitive to antimycin A. We therefore conclude that mitochondrial dysfunction activates vagal nociceptors by ROS-dependent (TRPA1) and ROS-independent (TRPV1) mechanisms.
Collapse
|
19
|
Nowak G, Bakajsova-Takacsova D. Protein kinase Cε targets respiratory chain and mitochondrial membrane potential but not F 0 F 1 -ATPase in renal cells injured by oxidant. J Cell Biochem 2018; 119:9394-9407. [PMID: 30074270 PMCID: PMC6298597 DOI: 10.1002/jcb.27256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/21/2018] [Indexed: 01/12/2023]
Abstract
We have previously shown that protein kinase Cε (PKCε) is involved in mitochondrial dysfunction in renal proximal tubular cells (RPTC). This study examined mitochondrial targets of active PKCε in RPTC injured by the model oxidant tert-butyl hydroperoxide (TBHP). TBHP exposure augmented the levels of phosphorylated (active) PKCε in mitochondria, which suggested translocation of PKCε to mitochondria after oxidant exposure. Oxidant injury decreased state 3 respiration, adenosine triphosphate (ATP) production, ATP content, and complex I activity. Further, TBHP exposure increased ΔΨm and production of reactive oxygen species (ROS), and induced mitochondrial fragmentation and RPTC death. PKCε activation by overexpressing constitutively active PKCε exacerbated decreases in state 3 respiration, complex I activity, ATP content, and augmented RPTC death. In contrast, inhibition of PKCε by overexpressing dnPKCε mutant restored state 3 respiration, respiratory control ratio, complex I activity, ΔΨm , and ATP production and content, but did not prevent decreases in F0 F1 -ATPase activity. Inhibition of PKCε prevented oxidant-induced production of ROS and mitochondrial fragmentation, and reduced RPTC death. We conclude that activation of PKCε mediates: (a) oxidant-induced changes in ΔΨm , decreases in mitochondrial respiration, complex I activity, and ATP content; (b) mitochondrial fragmentation; and (c) RPTC death. In contrast, oxidant-induced inhibition of F0 F1 -ATPase activity is not mediated by PKCε. These results show that, in contrast to the protective effects of PKCε in the heart, PKCε activation is detrimental to mitochondrial function and viability in RPTC and mediates oxidant-induced injury.
Collapse
Affiliation(s)
- Grazyna Nowak
- University of Arkansas for Medical Sciences, College of Pharmacy,
Department of Pharmaceutical Sciences, 4301 West Markham St., Little Rock, AR
72205
| | - Diana Bakajsova-Takacsova
- University of Arkansas for Medical Sciences, College of Pharmacy,
Department of Pharmaceutical Sciences, 4301 West Markham St., Little Rock, AR
72205
| |
Collapse
|
20
|
Metabolic Reprogramming and Redox Signaling in Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:241-260. [PMID: 29047090 DOI: 10.1007/978-3-319-63245-2_14] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pulmonary hypertension is a complex disease of the pulmonary vasculature, which in severe cases terminates in right heart failure. Complex remodeling of pulmonary arteries comprises the central issue of its pathology. This includes extensive proliferation, apoptotic resistance and inflammation. As such, the molecular and cellular features of pulmonary hypertension resemble hallmark characteristics of cancer cell behavior. The vascular remodeling derives from significant metabolic changes in resident cells, which we describe in detail. It affects not only cells of pulmonary artery wall, but also its immediate microenvironment involving cells of immune system (i.e., macrophages). Thus aberrant metabolism constitutes principle component of the cancer-like theory of pulmonary hypertension. The metabolic changes in pulmonary artery cells resemble the cancer associated Warburg effect, involving incomplete glucose oxidation through aerobic glycolysis with depressed mitochondrial catabolism enabling the fueling of anabolic reactions with amino acids, nucleotides and lipids to sustain proliferation. Macrophages also undergo overlapping but distinct metabolic reprogramming inducing specific activation or polarization states that enable their participation in the vascular remodeling process. Such metabolic synergy drives chronic inflammation further contributing to remodeling. Enhanced glycolytic flux together with suppressed mitochondrial bioenergetics promotes the accumulation of reducing equivalents, NAD(P)H. We discuss the enzymes and reactions involved. The reducing equivalents modulate the regulation of proteins using NAD(P)H as the transcriptional co-repressor C-terminal binding protein 1 cofactor and significantly impact redox status (through GSH, NAD(P)H oxidases, etc.), which together act to control the phenotype of the cells of pulmonary arteries. The altered mitochondrial metabolism changes its redox poise, which together with enhanced NAD(P)H oxidase activity and reduced enzymatic antioxidant activity promotes a pro-oxidative cellular status. Herein we discuss all described metabolic changes along with resultant alterations in redox status, which result in excessive proliferation, apoptotic resistance, and inflammation, further leading to pulmonary arterial wall remodeling and thus establishing pulmonary artery hypertension pathology.
Collapse
|
21
|
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: 4.0] [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
|
22
|
D'Alessandro A, El Kasmi KC, Plecitá-Hlavatá L, Ježek P, Li M, Zhang H, Gupte SA, Stenmark KR. Hallmarks of Pulmonary Hypertension: Mesenchymal and Inflammatory Cell Metabolic Reprogramming. Antioxid Redox Signal 2018; 28. [PMID: 28637353 PMCID: PMC5737722 DOI: 10.1089/ars.2017.7217] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE The molecular events that promote the development of pulmonary hypertension (PH) are complex and incompletely understood. The complex interplay between the pulmonary vasculature and its immediate microenvironment involving cells of immune system (i.e., macrophages) promotes a persistent inflammatory state, pathological angiogenesis, and fibrosis that are driven by metabolic reprogramming of mesenchymal and immune cells. Recent Advancements: Consistent with previous findings in the field of cancer metabolism, increased glycolytic rates, incomplete glucose and glutamine oxidation to support anabolism and anaplerosis, altered lipid synthesis/oxidation ratios, increased one-carbon metabolism, and activation of the pentose phosphate pathway to support nucleoside synthesis are but some of the key metabolic signatures of vascular cells in PH. In addition, metabolic reprogramming of macrophages is observed in PH and is characterized by distinct features, such as the induction of specific activation or polarization states that enable their participation in the vascular remodeling process. CRITICAL ISSUES Accumulation of reducing equivalents, such as NAD(P)H in PH cells, also contributes to their altered phenotype both directly and indirectly by regulating the activity of the transcriptional co-repressor C-terminal-binding protein 1 to control the proliferative/inflammatory gene expression in resident and immune cells. Further, similar to the role of anomalous metabolism in mitochondria in cancer, in PH short-term hypoxia-dependent and long-term hypoxia-independent alterations of mitochondrial activity, in the absence of genetic mutation of key mitochondrial enzymes, have been observed and explored as potential therapeutic targets. FUTURE DIRECTIONS For the foreseeable future, short- and long-term metabolic reprogramming will become a candidate druggable target in the treatment of PH. Antioxid. Redox Signal. 28, 230-250.
Collapse
Affiliation(s)
- Angelo D'Alessandro
- 1 Department of Biochemistry and Molecular Genetics, University of Colorado - Denver , Colorado
| | - Karim C El Kasmi
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado.,3 Department of Pediatric Gastroenterology, University of Colorado - Denver , Colorado
| | - Lydie Plecitá-Hlavatá
- 4 Department of Mitochondrial Physiology, Institute of Physiology , Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Ježek
- 4 Department of Mitochondrial Physiology, Institute of Physiology , Czech Academy of Sciences, Prague, Czech Republic
| | - Min Li
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| | - Hui Zhang
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| | - Sachin A Gupte
- 5 Department of Pharmacology, School of Medicine, New York Medical College , Valhalla, New York
| | - Kurt R Stenmark
- 2 Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado - Denver , Colorado
| |
Collapse
|
23
|
Song T, Zheng YM, Wang YX. Cross Talk Between Mitochondrial Reactive Oxygen Species and Sarcoplasmic Reticulum Calcium in Pulmonary Arterial Smooth Muscle Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:289-298. [DOI: 10.1007/978-3-319-63245-2_17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
24
|
Di Mise A, Wang YX, Zheng YM. Role of Transcription Factors in Pulmonary Artery Smooth Muscle Cells: An Important Link to Hypoxic Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:13-32. [PMID: 29047078 DOI: 10.1007/978-3-319-63245-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, namely a lack of oxygen in the blood, induces pulmonary vasoconstriction and vasoremodeling, which serve as essential pathologic factors leading to pulmonary hypertension (PH). The underlying molecular mechanisms are uncertain; however, pulmonary artery smooth muscle cells (PASMCs) play an essential role in hypoxia-induced pulmonary vasoconstriction, vasoremodeling, and PH. Hypoxia causes oxidative damage to DNAs, proteins, and lipids. This damage (oxidative stress) modulates the activity of ion channels and elevates the intracellular calcium concentration ([Ca2+]i, Ca2+ signaling) of PASMCs. The oxidative stress and increased Ca2+ signaling mutually interact with each other, and synergistically results in a variety of cellular responses. These responses include functional and structural abnormalities of mitochondria, sarcoplasmic reticulum, and nucleus; cell contraction, proliferation, migration, and apoptosis, as well as generation of vasoactive substances, inflammatory molecules, and growth factors that mediate the development of PH. A number of studies reveal that various transcription factors (TFs) play important roles in hypoxia-induced oxidative stress, disrupted PAMSC Ca2+ signaling and the development and progress of PH. It is believed that in the pathogenesis of PH, hypoxia facilitates these roles by mediating the expression of multiple genes. Therefore, the identification of specific genes and their transcription factors implicated in PH is necessary for the complete understanding of the underlying molecular mechanisms. Moreover, this identification may aid in the development of novel and effective therapeutic strategies for PH.
Collapse
Affiliation(s)
- Annarita Di Mise
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| | - Yun-Min Zheng
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
25
|
Ringvold HC, Khalil RA. Protein Kinase C as Regulator of Vascular Smooth Muscle Function and Potential Target in Vascular Disorders. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:203-301. [PMID: 28212798 PMCID: PMC5319769 DOI: 10.1016/bs.apha.2016.06.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vascular smooth muscle (VSM) plays an important role in maintaining vascular tone. In addition to Ca2+-dependent myosin light chain (MLC) phosphorylation, protein kinase C (PKC) is a major regulator of VSM function. PKC is a family of conventional Ca2+-dependent α, β, and γ, novel Ca2+-independent δ, ɛ, θ, and η, and atypical ξ, and ι/λ isoforms. Inactive PKC is mainly cytosolic, and upon activation it undergoes phosphorylation, maturation, and translocation to the surface membrane, the nucleus, endoplasmic reticulum, and other cell organelles; a process facilitated by scaffold proteins such as RACKs. Activated PKC phosphorylates different substrates including ion channels, pumps, and nuclear proteins. PKC also phosphorylates CPI-17 leading to inhibition of MLC phosphatase, increased MLC phosphorylation, and enhanced VSM contraction. PKC could also initiate a cascade of protein kinases leading to phosphorylation of the actin-binding proteins calponin and caldesmon, increased actin-myosin interaction, and VSM contraction. Increased PKC activity has been associated with vascular disorders including ischemia-reperfusion injury, coronary artery disease, hypertension, and diabetic vasculopathy. PKC inhibitors could test the role of PKC in different systems and could reduce PKC hyperactivity in vascular disorders. First-generation PKC inhibitors such as staurosporine and chelerythrine are not very specific. Isoform-specific PKC inhibitors such as ruboxistaurin have been tested in clinical trials. Target delivery of PKC pseudosubstrate inhibitory peptides and PKC siRNA may be useful in localized vascular disease. Further studies of PKC and its role in VSM should help design isoform-specific PKC modulators that are experimentally potent and clinically safe to target PKC in vascular disease.
Collapse
Affiliation(s)
- H C Ringvold
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - R A Khalil
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
26
|
Waypa GB, Smith KA, Schumacker PT. O2 sensing, mitochondria and ROS signaling: The fog is lifting. Mol Aspects Med 2016; 47-48:76-89. [PMID: 26776678 DOI: 10.1016/j.mam.2016.01.002] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 12/14/2022]
Abstract
Mitochondria are responsible for the majority of oxygen consumption in cells, and thus represent a conceptually appealing site for cellular oxygen sensing. Over the past 40 years, a number of mechanisms to explain how mitochondria participate in oxygen sensing have been proposed. However, no consensus has been reached regarding how mitochondria could regulate transcriptional and post-translational responses to hypoxia. Nevertheless, a growing body of data continues to implicate a role for increased reactive oxygen species (ROS) signals from the electron transport chain (ETC) in triggering responses to hypoxia in diverse cell types. The present article reviews our progress in understanding this field and considers recent advances that provide new insight, helping to lift the fog from this complex topic.
Collapse
Affiliation(s)
- Gregory B Waypa
- Department of Pediatrics, Division of Neonatology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Kimberly A Smith
- Department of Pediatrics, Division of Neonatology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Paul T Schumacker
- Department of Pediatrics, Division of Neonatology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
27
|
Sommer N, Strielkov I, Pak O, Weissmann N. Oxygen sensing and signal transduction in hypoxic pulmonary vasoconstriction. Eur Respir J 2015; 47:288-303. [PMID: 26493804 DOI: 10.1183/13993003.00945-2015] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/24/2015] [Indexed: 01/17/2023]
Abstract
Hypoxic pulmonary vasoconstriction (HPV), also known as the von Euler-Liljestrand mechanism, is an essential response of the pulmonary vasculature to acute and sustained alveolar hypoxia. During local alveolar hypoxia, HPV matches perfusion to ventilation to maintain optimal arterial oxygenation. In contrast, during global alveolar hypoxia, HPV leads to pulmonary hypertension. The oxygen sensing and signal transduction machinery is located in the pulmonary arterial smooth muscle cells (PASMCs) of the pre-capillary vessels, albeit the physiological response may be modulated in vivo by the endothelium. While factors such as nitric oxide modulate HPV, reactive oxygen species (ROS) have been suggested to act as essential mediators in HPV. ROS may originate from mitochondria and/or NADPH oxidases but the exact oxygen sensing mechanisms, as well as the question of whether increased or decreased ROS cause HPV, are under debate. ROS may induce intracellular calcium increase and subsequent contraction of PASMCs via direct or indirect interactions with protein kinases, phospholipases, sarcoplasmic calcium channels, transient receptor potential channels, voltage-dependent potassium channels and L-type calcium channels, whose relevance may vary under different experimental conditions. Successful identification of factors regulating HPV may allow development of novel therapeutic approaches for conditions of disturbed HPV.
Collapse
Affiliation(s)
- Natascha Sommer
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Ievgen Strielkov
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Oleg Pak
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System, University of Giessen Lung Center, German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
28
|
Abstract
SIGNIFICANCE The pulmonary circulation is a low-pressure, low-resistance, highly compliant vasculature. In contrast to the systemic circulation, it is not primarily regulated by a central nervous control mechanism. The regulation of resting membrane potential due to ion channels is of integral importance in the physiology and pathophysiology of the pulmonary vasculature. RECENT ADVANCES Redox-driven ion conductance changes initiated by direct oxidation, nitration, and S-nitrosylation of the cysteine thiols and indirect phosphorylation of the threonine and serine residues directly affect pulmonary vascular tone. CRITICAL ISSUES Molecular mechanisms of changes in ion channel conductance, especially the identification of the sites of action, are still not fully elucidated. FUTURE DIRECTIONS Further investigation of the interaction between redox status and ion channel gating, especially the physiological significance of S-glutathionylation and S-nitrosylation, could result in a better understanding of the physiological and pathophysiological importance of these mediators in general and the implications of such modifications in cellular functions and related diseases and their importance for targeted treatment strategies.
Collapse
Affiliation(s)
- Andrea Olschewski
- 1 Ludwig Boltzmann Institute for Lung Vascular Research , Graz, Austria
| | | |
Collapse
|
29
|
Xiao D, Hu XQ, Huang X, Zhou J, Wilson SM, Yang S, Zhang L. Chronic hypoxia during gestation enhances uterine arterial myogenic tone via heightened oxidative stress. PLoS One 2013; 8:e73731. [PMID: 24066066 PMCID: PMC3774750 DOI: 10.1371/journal.pone.0073731] [Citation(s) in RCA: 34] [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: 05/10/2013] [Accepted: 07/19/2013] [Indexed: 11/25/2022] Open
Abstract
Chronic hypoxia during gestation has profound adverse effects on the adaptation of uteroplacental circulation in pregnancy. Yet, the underlying mechanisms are not fully understood. The present study tested the hypothesis that enhanced production of reactive oxygen species (ROS) in uterine arteries plays a critical role in the maladaptation of uterine circulation associated with chronic hypoxia. Uterine arteries were isolated from nonpregnant and near-term pregnant sheep maintained at sea level (~300 m) or exposed to high-altitude (3801 m) hypoxia for 110 days. Hypoxia significantly increased ROS production in uterine arteries of pregnant, but not nonpregnant, sheep. This was associated with a significant increase in NADPH oxidase (Nox) 2, but not Nox1 or Nox4, protein abundance and total Nox activity in uterine arteries of pregnant animals. Chronic hypoxia significantly increased pressure-dependent uterine arterial myogenic tone in pregnant sheep, which was abrogated by a Nox inhibitor apocynin. Additionally, the hypoxia-induced increase in myogenic reactivity of uterine arteries to phorbol 12,13-dibutyrate in pregnant sheep was blocked by apocynin and tempol. In consistence with the myogenic responses, the hypoxia-mediated down-regulation of BKCa channel activity in uterine arteries of pregnant animals was reversed by apocynin. The findings suggest that heightened oxidative stress in uterine arteries plays a key role in suppressing the BKCa channel activity, resulting in increased myogenic reactivity and maladaptation of uteroplacental circulation caused by chronic hypoxia during gestation.
Collapse
Affiliation(s)
- Daliao Xiao
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Xiang-Qun Hu
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Xiaohui Huang
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Jianjun Zhou
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Sean M. Wilson
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| | - Shumei Yang
- Department of Chemistry and Biochemistry, California State University, San Bernardino, California, United States of America
| | - Lubo Zhang
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, United States of America
| |
Collapse
|
30
|
Tuo QR, Ma YF, Chen W, Luo XJ, Shen J, Guo D, Zheng YM, Wang YX, Ji G, Liu QH. Reactive oxygen species induce a Ca(2+)-spark increase in sensitized murine airway smooth muscle cells. Biochem Biophys Res Commun 2013; 434:498-502. [PMID: 23583396 DOI: 10.1016/j.bbrc.2013.03.102] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 03/26/2013] [Indexed: 10/27/2022]
Abstract
The level of reactive oxygen species (ROS) and the activity of spontaneous, transient, localized Ca(2+) increases (known as Ca(2+) sparks) in tracheal smooth muscle cells (TSMCs) in an experimental allergic asthma mouse model has not yet been investigated. We used laser confocal microscopy and fluorescent dyes to measure ROS levels and Ca(2+) sparks, and we found that both events were significantly increased in TSMCs obtained from ovalbumin (OVA)-sensitized/-challenged mice compared with control mice. ROS levels began to increase in TSMCs after the first OVA challenge, and this increase was sustained. However, this elevation and Ca(2+)-spark increase was abolished after the administration of the ROS scavenger N-acetylcysteine amide (NACA) for 5days. Furthermore, a similar inhibition was also observed following the direct perfusion of NACA into cells isolated from the (OVA)-sensitized mice that were not treated with NACA. Moreover, we used 0.1-mM caffeine treatment to increase the Ca(2+) sparks in single TSMCs and observed cell shortening. In addition, we did not find increases in the mRNA levels of ryanodine (RyRs) and inositol 1,4,5-trisphosphate (IP3Rs) receptors in the tracheal smooth muscle cells of (OVA)-sensitized mice compared with controls. We concluded that ROS and Ca(2+) sparks increased in (OVA)-sensitized TSMCs. We found that ROS induces Ca(2+) sparks, and increased Ca(2+) sparks resulted in the contraction of (OVA)-sensitized TSMCs, resulting in the generation of airway hyperresponsiveness (AHR). This effect may represent a novel mechanism for AHR pathogenesis and might provide insight into new methods for the clinical prevention and treatment of asthma and asthmatic AHR.
Collapse
Affiliation(s)
- Qing-Rong Tuo
- Institute for Medical Biology, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Strielkov I, Kizub I, Khromov A, Soloviev A. Evidence for the role of phosphatidylcholine-specific phospholipase C in sustained hypoxic pulmonary vasoconstriction. Vascul Pharmacol 2013; 58:292-8. [DOI: 10.1016/j.vph.2013.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/31/2013] [Accepted: 02/04/2013] [Indexed: 11/29/2022]
|
32
|
Puri N, Zhang F, Monu SR, Sodhi K, Bellner L, Lamon BD, Zhang Y, Abraham NG, Nasjletti A. Antioxidants condition pleiotropic vascular responses to exogenous H(2)O(2): role of modulation of vascular TP receptors and the heme oxygenase system. Antioxid Redox Signal 2013; 18:471-80. [PMID: 22867102 PMCID: PMC3545357 DOI: 10.1089/ars.2012.4587] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 08/06/2012] [Accepted: 08/07/2012] [Indexed: 11/13/2022]
Abstract
AIMS Hydrogen peroxide (H(2)O(2)), a nonradical oxidant, is employed to ascertain the role of redox mechanisms in regulation of vascular tone. Where both dilation and constriction have been reported, we examined the hypothesis that the ability of H(2)O(2) to effect vasoconstriction or dilation is conditioned by redox mechanisms and may be modulated by antioxidants. RESULTS Exogenous H(2)O(2) (0.1-10.0 μM), dose-dependently reduced the internal diameter of rat renal interlobular and 3rd-order mesenteric arteries (p<0.05). This response was obliterated in arteries pretreated with antioxidants, including tempol, pegylated superoxide dismutase (PEG-SOD), butylated hydroxytoluene (BHT), and biliverdin (BV). However, as opposed to tempol or PEG-SOD, BHT & BV, antioxidants targeting radicals downstream of H(2)O(2), also uncovered vasodilation. INNOVATIONS Redox-dependent vasoconstriction to H(2)O(2) was blocked by inhibitors of cyclooxygenase (COX) (indomethacin-10 μM), thromboxane (TP) synthase (CGS13080-10 μM), and TP receptor antagonist (SQ29548-1 μM). However, H(2)O(2) did not increase vascular thromboxane B(2) release; instead, it sensitized the vasculature to a TP agonist, U46619, an effect reversed by PEG-SOD. Antioxidant-conditioned dilatory response to H(2)O(2) was accompanied by enhanced vascular heme oxygenase (HO)-dependent carbon monoxide generation and was abolished by HO inhibitors or by HO-1 & 2 antisense oligodeoxynucleotides treatment of SD rats. CONCLUSION These results demonstrate that H(2)O(2) has antioxidant-modifiable pleiotropic vascular effects, where constriction and dilation are brought about in the same vascular segment. H(2)O(2)-induced oxidative stress increases vascular TP sensitivity and predisposes these arterial segments to constrictor prostanoids. Conversely, vasodilation is reliant upon HO-derived products whose synthesis is stimulated only in the presence of antioxidants targeting radicals downstream of H(2)O(2).
Collapse
Affiliation(s)
- Nitin Puri
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Freund-Michel V, Guibert C, Dubois M, Courtois A, Marthan R, Savineau JP, Muller B. Reactive oxygen species as therapeutic targets in pulmonary hypertension. Ther Adv Respir Dis 2013; 7:175-200. [PMID: 23328248 DOI: 10.1177/1753465812472940] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pulmonary hypertension (PH) is characterized by a progressive elevation of pulmonary arterial pressure due to alterations of both pulmonary vascular structure and function. This disease is rare but life-threatening, leading to the development of right heart failure. Current PH treatments, designed to target altered pulmonary vascular reactivity, include vasodilating prostanoids, phosphodiesterase-5 inhibitors and endothelin-1 receptor antagonists. Although managing to slow the progression of the disease, these molecules still do not cure PH. More effective treatments need to be developed, and novel therapeutic strategies, targeting in particular vascular remodelling, are currently under investigation. Reactive oxygen species (ROS) are important physiological messengers in vascular cells. In addition to atherosclerosis and other systemic vascular diseases, emerging evidence also support a role of ROS in PH pathogenesis. ROS production is increased in animal models of PH, associated with NADPH oxidases increased expression, in particular of several Nox enzymes thought to be the major source of ROS in the pulmonary vasculature. These increases have also been observed in vitro and in vivo in humans. Moreover, several studies have shown either the deleterious effect of agents promoting ROS generation on pulmonary vasculature or, conversely, the beneficial effect of antioxidant agents in animal models of PH. In these studies, ROS production has been directly linked to pulmonary vascular remodelling, endothelial dysfunction, altered vasoconstrictive responses, inflammation and modifications of the extracellular matrix, all important features of PH pathophysiology. Altogether, these findings indicate that ROS are interesting therapeutic targets in PH. Blockade of ROS-dependent signalling pathways, or disruption of sources of ROS in the pulmonary vasculature, targeting in particular Nox enzymes, represent promising new therapeutic strategies in this disease.
Collapse
Affiliation(s)
- Véronique Freund-Michel
- Laboratoire de Pharmacologie-INSERM U1045, UFR des Sciences Pharmaceutiques, Université Bordeaux Segalen, Case 83, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France.
| | | | | | | | | | | | | |
Collapse
|
34
|
Yadav VR, Song T, Joseph L, Mei L, Zheng YM, Wang YX. Important role of PLC-γ1 in hypoxic increase in intracellular calcium in pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2012. [PMID: 23204067 DOI: 10.1152/ajplung.00310.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An increase in intracellular calcium concentration ([Ca(2+)](i)) in pulmonary arterial smooth muscle cells (PASMCs) induces hypoxic cellular responses in the lungs; however, the underlying molecular mechanisms remain incompletely understood. We report, for the first time, that acute hypoxia significantly enhances phospholipase C (PLC) activity in mouse resistance pulmonary arteries (PAs), but not in mesenteric arteries. Western blot analysis and immunofluorescence staining reveal the expression of PLC-γ1 protein in PAs and PASMCs, respectively. The activity of PLC-γ1 is also augmented in PASMCs following hypoxia. Lentiviral shRNA-mediated gene knockdown of mitochondrial complex III Rieske iron-sulfur protein (RISP) to inhibit reactive oxygen species (ROS) production prevents hypoxia from increasing PLC-γ1 activity in PASMCs. Myxothiazol, a mitochondrial complex III inhibitor, reduces the hypoxic response as well. The PLC inhibitor U73122, but not its inactive analog U73433, attenuates the hypoxic vasoconstriction in PAs and hypoxic increase in [Ca(2+)](i) in PASMCs. PLC-γ1 knockdown suppresses its protein expression and the hypoxic increase in [Ca(2+)](i). Hypoxia remarkably increases inositol 1,4,5-trisphosphate (IP(3)) production, which is blocked by U73122. The IP(3) receptor (IP(3)R) antagonist 2-aminoethoxydiphenyl borate (2-APB) or xestospongin-C inhibits the hypoxic increase in [Ca(2+)](i). PLC-γ1 knockdown or U73122 reduces H(2)O(2)-induced increase in [Ca(2+)](i) in PASMCs and contraction in PAs. 2-APB and xestospongin-C produce similar inhibitory effects. In conclusion, our findings provide novel evidence that hypoxia activates PLC-γ1 by increasing RISP-dependent mitochondrial ROS production in the complex III, which causes IP(3) production, IP(3)R opening, and Ca(2+) release, playing an important role in hypoxic Ca(2+) and contractile responses in PASMCs.
Collapse
Affiliation(s)
- Vishal R Yadav
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Cosentino-Gomes D, Rocco-Machado N, Meyer-Fernandes JR. Cell signaling through protein kinase C oxidation and activation. Int J Mol Sci 2012; 13:10697-10721. [PMID: 23109817 PMCID: PMC3472709 DOI: 10.3390/ijms130910697] [Citation(s) in RCA: 187] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 08/02/2012] [Accepted: 08/13/2012] [Indexed: 01/15/2023] Open
Abstract
Due to the growing importance of cellular signaling mediated by reactive oxygen species (ROS), proteins that are reversibly modulated by these reactant molecules are of high interest. In this context, protein kinases and phosphatases, which act coordinately in the regulation of signal transduction through the phosphorylation and dephosphorylation of target proteins, have been described to be key elements in ROS-mediated signaling events. The major mechanism by which these proteins may be modified by oxidation involves the presence of key redox-sensitive cysteine residues. Protein kinase C (PKC) is involved in a variety of cellular signaling pathways. These proteins have been shown to contain a unique structural feature that is susceptible to oxidative modification. A large number of scientific studies have highlighted the importance of ROS as a second messenger in numerous cellular processes, including cell proliferation, gene expression, adhesion, differentiation, senescence, and apoptosis. In this context, the goal of this review is to discuss the mechanisms by which PKCs are modulated by ROS and how these processes are involved in the cellular response.
Collapse
Affiliation(s)
- Daniela Cosentino-Gomes
- Institute of Medical Biochemistry, Federal University of Rio de Janeiro (UFRJ), CCS, Bloco H, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil; E-Mails: (N.R.-M.); (J.R.M.-F.)
- Institute of National Science and Technology of Structural Biology and Bioimage (INCTBEB), CCS, Bloco H, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +55-21-2562-6781; Fax: +55-21-2270-8647
| | - Nathália Rocco-Machado
- Institute of Medical Biochemistry, Federal University of Rio de Janeiro (UFRJ), CCS, Bloco H, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil; E-Mails: (N.R.-M.); (J.R.M.-F.)
- Institute of National Science and Technology of Structural Biology and Bioimage (INCTBEB), CCS, Bloco H, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil
| | - José Roberto Meyer-Fernandes
- Institute of Medical Biochemistry, Federal University of Rio de Janeiro (UFRJ), CCS, Bloco H, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil; E-Mails: (N.R.-M.); (J.R.M.-F.)
- Institute of National Science and Technology of Structural Biology and Bioimage (INCTBEB), CCS, Bloco H, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
36
|
Effect of Yifei Huoxue Granule on the proliferation of rat pulmonary artery smooth muscle cells upon exposure to chronic hypoxic conditions in vitro. Chin J Integr Med 2012; 18:507-13. [PMID: 22772913 DOI: 10.1007/s11655-012-1150-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Indexed: 10/28/2022]
Abstract
OBJECTIVE To investigate the inhibitory effect of Yifei Huoxue Granule (, YFHXG) on the hypoxia-induced proliferation of rat pulmonary artery smooth muscle cells (PASMCs) and its mechanism of decreasing pulmonary arterial pressure. METHODS Twenty male Sprague-Dawley (SD) rats were randomly divided into four groups: saline, and 0.66, 3.30 and 16.50 g/kg of YFHXG groups, the saline and different concentrations of YFHXG were given twice daily for 7 days, respectively. Serum-pharmacology method was used in the preparation of YFHXG serum. Tissue block anchorage was employed in the primary culture of rat PASMCs. The PASMCs were randomly divided into normoxia group, hypoxia group, and hypoxia+YFHXG group (0.66, 3.30 and 16.50 g/kg doses of YFHXG-treated serum groups, exposed to hypoxic condition). PASMCs in normoxia and hypoxia group were cultured with saline serum, hypoxia+YFHXG groups were cultured with different concentrations of YFHXG serum. Cell viability was assessed with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell cycle was analyzed using flow cytometry. In addition, hypoxia inducible factor-1-alpha (HIF-1α) protein expression was evaluated by immunocytochemistry analysis, the concentration of intracellular reactive oxygen species (ROS) and Ca(2+) were determined by laser scanning confocal microscopy (LSCM). RESULTS MTT assay and flow cytometry showed that hypoxia could directly activate the proliferation of PASMCs, while YFHXG dose-dependently inhibited hypoxia-induced proliferation of rat PASMCs. Immunocytochemistry showed that hypoxia enhanced HIF-1α protein expression, and LSCM showed that hypoxia significantly increased intracellular ROS and Ca(2+), while YFHXG decreased the expression of HIF- 1α and attenuated the hypoxia-induced increase in intracellular concentration of ROS and Ca(2+). CONCLUSIONS YFHXG could inhibit hypoxia-induced proliferation of rat PASMCs, which may decrease pulmonary arterial pressure and vascular remodeling. The anti-hypoxia effect of YFHXG may be explained by its regulation of HIF-1α expression and of the levels of intracellular ROS and Ca(2+).
Collapse
|
37
|
Frazziano G, Champion HC, Pagano PJ. NADPH oxidase-derived ROS and the regulation of pulmonary vessel tone. Am J Physiol Heart Circ Physiol 2012; 302:H2166-77. [PMID: 22427511 DOI: 10.1152/ajpheart.00780.2011] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary vessel constriction results from an imbalance between vasodilator and vasoconstrictor factors released by the endothelium including nitric oxide, endothelin, prostanoids, and reactive oxygen species (ROS). ROS, generated by a variety of enzymatic sources (such as mitochondria and NADPH oxidases, a.k.a. Nox), appear to play a pivotal role in vascular homeostasis, whereas elevated levels effect vascular disease. The pulmonary circulation is very sensitive to changes in the partial pressure of oxygen and differs from the systemic circulation in its response to this change. In fact, the pulmonary vessels contract in response to low oxygen tension, whereas systemic vessels dilate. Growing evidence suggests that ROS production and ROS-related pathways may be key factors that underlie this differential response to oxygen tension. A major emphasis of our laboratory is the role of Nox isozymes in cardiovascular disease. In this review, we will focus our attention on the role of Nox-derived ROS in the control of pulmonary vascular tone.
Collapse
Affiliation(s)
- G Frazziano
- Department of Pharmacology and Chemical Biology and Vascular Medicine Institute, University of Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
38
|
Abstract
It has been known for more than 60 years, and suspected for over 100, that alveolar hypoxia causes pulmonary vasoconstriction by means of mechanisms local to the lung. For the last 20 years, it has been clear that the essential sensor, transduction, and effector mechanisms responsible for hypoxic pulmonary vasoconstriction (HPV) reside in the pulmonary arterial smooth muscle cell. The main focus of this review is the cellular and molecular work performed to clarify these intrinsic mechanisms and to determine how they are facilitated and inhibited by the extrinsic influences of other cells. Because the interaction of intrinsic and extrinsic mechanisms is likely to shape expression of HPV in vivo, we relate results obtained in cells to HPV in more intact preparations, such as intact and isolated lungs and isolated pulmonary vessels. Finally, we evaluate evidence regarding the contribution of HPV to the physiological and pathophysiological processes involved in the transition from fetal to neonatal life, pulmonary gas exchange, high-altitude pulmonary edema, and pulmonary hypertension. Although understanding of HPV has advanced significantly, major areas of ignorance and uncertainty await resolution.
Collapse
Affiliation(s)
- J T Sylvester
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, The Johns Hopkins University School ofMedicine, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
39
|
Frazziano G, Moreno L, Moral-Sanz J, Menendez C, Escolano L, Gonzalez C, Villamor E, Alvarez-Sala JL, Cogolludo AL, Perez-Vizcaino F. Neutral sphingomyelinase, NADPH oxidase and reactive oxygen species. Role in acute hypoxic pulmonary vasoconstriction. J Cell Physiol 2011; 226:2633-40. [PMID: 21792922 DOI: 10.1002/jcp.22611] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The molecular mechanisms underlying hypoxic pulmonary vasoconstriction (HPV) are not yet properly understood. Mitochondrial electron transport chain (ETC) and NADPH oxidase have been proposed as possible oxygen sensors, with derived reactive oxygen species (ROS) playing key roles in coupling the sensor(s) to the contractile machinery. We have recently reported that activation of neutral sphingomyelinase (nSMase) and protein kinase C ζ (PKCζ) participate in the signalling cascade of HPV. Herein, we studied the significance of nSMase in controlling ROS production rate in rat pulmonary artery (PA) smooth muscle cells and thereby HPV in rat PA. ROS production (analyzed by dichlorofluorescein and dihydroethidium fluorescence) was increased by hypoxia in endothelium-denuded PA segments and their inhibition prevented hypoxia-induced voltage-gated potassium channel (K(V) ) inhibition and pulmonary vasoconstriction. Consistently, H(2) O(2) , or its analogue t-BHP, decreased K(V) currents and induced a contractile response, mimicking the effects of hypoxia. Inhibitors of mitochondrial ETC (rotenone) and NADPH oxidase (apocynin) prevented hypoxia-induced ROS production, K(V) channel inhibition and vasoconstriction. Hypoxia induced p47(phox) phosphorylation and its interaction with caveolin-1. Inhibition of nSMase (GW4869) or PKCζ prevented p47(phox) phosphorylation and ROS production. The increase in ceramide induced by hypoxia (analyzed by immunocytochemistry) was inhibited by rotenone. Exogenous ceramide increased ROS production in a PKCζ sensitive manner. We propose an integrated signalling pathway for HPV which includes nSMase-PKCζ-NADPH oxidase as a necessary step required for ROS production and vasoconstriction.
Collapse
Affiliation(s)
- Giovanna Frazziano
- Department of Pharmacology, School of Medicine, University Complutense of Madrid, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Knock GA, Ward JPT. Redox regulation of protein kinases as a modulator of vascular function. Antioxid Redox Signal 2011; 15:1531-47. [PMID: 20849377 DOI: 10.1089/ars.2010.3614] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Reactive oxygen species (ROS) are continuously generated in vascular tissues by various oxidoreductase enzymes. They contribute to normal cell signaling, and modulate vascular smooth muscle tone and endothelial permeability in response to physiological agonists and to various cellular stresses and environmental factors, such as hypoxia. While concentrations of ROS are normally tightly controlled by cellular redox buffer systems, if produced in excess they may contribute to vascular disease. Protein kinases are essential components of most cell signaling pathways, including those involving ROS. The functioning of several members of this highly diverse group of enzymes, which include receptor and nonreceptor tyrosine kinases, protein kinase C, mitogen-activated kinases, and Rho-kinase, are modified by ROS, either through direct oxidative modification or indirectly through modification of associated proteins such as tyrosine phosphatases and monomeric G proteins. In this review, we discuss the molecular mechanisms of redox modification of these proteins, the downstream pathways affected, the often complex interaction between major kinase pathways, and feedback to ROS production itself. We also discuss complicating factors such as differential actions of superoxide anion and hydrogen peroxide, questions concerning concentration dependence, and the significance of signaling microdomains.
Collapse
Affiliation(s)
- Greg A Knock
- Division of Asthma, Allergy, and Lung Biology, King's College London, Stamford Street, London, United Kingdom.
| | | |
Collapse
|
41
|
Korde AS, Yadav VR, Zheng YM, Wang YX. Primary role of mitochondrial Rieske iron-sulfur protein in hypoxic ROS production in pulmonary artery myocytes. Free Radic Biol Med 2011; 50:945-52. [PMID: 21238580 PMCID: PMC3051030 DOI: 10.1016/j.freeradbiomed.2011.01.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 01/04/2011] [Accepted: 01/06/2011] [Indexed: 12/20/2022]
Abstract
This study was designed to determine whether: (1) hypoxia could directly affect ROS production in isolated mitochondria and mitochondrial complex III from pulmonary artery smooth muscle cells (PASMCs) and (2) Rieske iron-sulfur protein in complex III might mediate hypoxic ROS production, leading to hypoxic pulmonary vasoconstriction (HPV). Our data, for the first time, demonstrate that hypoxia significantly enhances ROS production, measured by the standard ROS indicator dichlorodihydrofluorescein/diacetate, in isolated mitochondria from PASMCs. Studies using the newly developed, specific ROS biosensor pHyPer have found that hypoxia increases mitochondrial ROS generation in isolated PASMCs as well. Hypoxic ROS production has also been observed in isolated complex III. Rieske iron-sulfur protein silencing using siRNA abolishes the hypoxic ROS formation in isolated PASM complex III, mitochondria, and cells, whereas Rieske iron-sulfur protein overexpression produces the opposite effect. Rieske iron-sulfur protein silencing inhibits the hypoxic increase in [Ca(2+)](i) in PASMCs and hypoxic vasoconstriction in isolated PAs. These findings together provide novel evidence that mitochondria are the direct hypoxic targets in PASMCs, in which Rieske iron-sulfur protein in complex III may serve as an essential, primary molecule that mediates the hypoxic ROS generation, leading to an increase in intracellular Ca(2+) in PASMCs and HPV.
Collapse
Affiliation(s)
| | | | | | - Yong-Xiao Wang
- Corresponding author: Dr. Yong-Xiao Wang Albany Medical College Center for Cardiovascular Sciences (MC-8) 47 New Scotland Avenue Albany, NY 12208 Phone: (518)-262-9506 Fax: (518)-262-8101
| |
Collapse
|
42
|
Hypoxia-induced sensitization of transient receptor potential vanilloid 1 involves activation of hypoxia-inducible factor-1 alpha and PKC. Pain 2011; 152:936-945. [DOI: 10.1016/j.pain.2011.02.024] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 12/26/2010] [Accepted: 02/07/2011] [Indexed: 02/01/2023]
|
43
|
Liao B, Zheng YM, Yadav VR, Korde AS, Wang YX. Hypoxia induces intracellular Ca2+ release by causing reactive oxygen species-mediated dissociation of FK506-binding protein 12.6 from ryanodine receptor 2 in pulmonary artery myocytes. Antioxid Redox Signal 2011; 14:37-47. [PMID: 20518593 PMCID: PMC3000638 DOI: 10.1089/ars.2009.3047] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 05/08/2010] [Accepted: 06/02/2010] [Indexed: 11/12/2022]
Abstract
Here we attempted to test a novel hypothesis that hypoxia may induce Ca(2+) release through reactive oxygen species (ROS)-mediated dissociation of FK506-binding protein 12.6 (FKBP12.6) from ryanodine receptors (RyRs) on the sarcoplasmic reticulum (SR) in pulmonary artery smooth muscle cells (PASMCs). The results reveal that hypoxic exposure significantly decreased the amount of FKBP12.6 on the SR of PAs and increased FKBP12.6 in the cytosol. The colocalization of FKBP12.6 with RyRs was decreased in intact PASMCs. Pharmacological and genetic inhibition of intracellular ROS generation prevented hypoxia from decreasing FKBP12.6 on the SR and increasing FKBP12.6 in the cytosol. Exogenous ROS (H(2)O(2)) reduced FKBP12.6 on the SR and augmented FKBP12.6 in the cytosol. Oxidized FKBP12.6 was absent on the SR from PAs pretreated with and without hypoxia, but it was present with a higher amount in the cytosol from PAs pretreated with than without hypoxia. Hypoxia and H(2)O(2) diminished the association of FKBP12.6 from type 2 RyRs (RyR2). The activity of RyRs was increased in PAs pretreated with hypoxia or H(2)O(2). FKBP12.6 removal enhanced, whereas RyR2 gene deletion blocked the hypoxic increase in [Ca(2+)](i) in PASMCs. Collectively, we conclude that hypoxia may induce Ca(2+) release by causing ROS-mediated dissociation of FKBP12.6 from RyR2 in PASMCs.
Collapse
Affiliation(s)
- Bo Liao
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York, USA
| | | | | | | | | |
Collapse
|
44
|
Gonzalez C, Agapito MT, Rocher A, Gomez-Niño A, Rigual R, Castañeda J, Conde SV, Obeso A. A revisit to O2 sensing and transduction in the carotid body chemoreceptors in the context of reactive oxygen species biology. Respir Physiol Neurobiol 2010; 174:317-30. [PMID: 20833275 DOI: 10.1016/j.resp.2010.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2010] [Revised: 08/31/2010] [Accepted: 09/01/2010] [Indexed: 11/25/2022]
Abstract
Oxygen-sensing and transduction in purposeful responses in cells and organisms is of great physiological and medical interest. All animals, including humans, encounter in their lifespan many situations in which oxygen availability might be insufficient, whether acutely or chronically, physiologically or pathologically. Therefore to trace at the molecular level the sequence of events or steps connecting the oxygen deficit with the cell responses is of interest in itself as an achievement of science. In addition, it is also of great medical interest as such knowledge might facilitate the therapeutical approach to patients and to design strategies to minimize hypoxic damage. In our article we define the concepts of sensors and transducers, the steps of the hypoxic transduction cascade in the carotid body chemoreceptor cells and also discuss current models of oxygen- sensing (bioenergetic, biosynthetic and conformational) with their supportive and unsupportive data from updated literature. We envision oxygen-sensing in carotid body chemoreceptor cells as a process initiated at the level of plasma membrane and performed by a hemoprotein, which might be NOX4 or a hemoprotein not yet chemically identified. Upon oxygen-desaturation, the sensor would experience conformational changes allosterically transmitted to oxygen regulated K+ channels, the initial effectors in the transduction cascade. A decrease in their opening probability would produce cell depolarization, activation of voltage dependent calcium channels and release of neurotransmitters. Neurotransmitters would activate the nerve endings of the carotid body sensory nerve to convey the information of the hypoxic situation to the central nervous system that would command ventilation to fight hypoxia.
Collapse
Affiliation(s)
- C Gonzalez
- Departamento de Bioquímica y Biología Molecular y Fisiología, Instituto de Biología y Genética Molecular y CIBER de Enfermedades Respiratorias, Universidad de Valladolid, Consejo Superior de Investigaciones Científicas e Instituto Carlos III, Facultad de Medicina, 47005 Valladolid, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Interactions between calcium and reactive oxygen species in pulmonary arterial smooth muscle responses to hypoxia. Respir Physiol Neurobiol 2010; 174:221-9. [PMID: 20801238 DOI: 10.1016/j.resp.2010.08.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 08/18/2010] [Accepted: 08/20/2010] [Indexed: 02/07/2023]
Abstract
In contrast to the systemic vasculature, where hypoxia causes vasodilation, pulmonary arteries constrict in response to hypoxia. The mechanisms underlying this unique response have been the subject of investigation for over 50 years, and still remain a topic of great debate. Over the last 20 years, there has emerged a general consensus that both increases in intracellular calcium concentration and changes in reactive oxygen species (ROS) generation play key roles in the pulmonary vascular response to hypoxia. Controversy exists, however, regarding whether ROS increase or decrease during hypoxia, the source of ROS, and the mechanisms by which changes in ROS might impact intracellular calcium, and vice versa. This review will discuss the mechanisms regulating [Ca2+]i and ROS in PASMCs, and the interaction between ROS and Ca2+ signaling during exposure to acute hypoxia.
Collapse
|
46
|
Perez-Vizcaino F, Cogolludo A, Moreno L. Reactive oxygen species signaling in pulmonary vascular smooth muscle. Respir Physiol Neurobiol 2010; 174:212-20. [PMID: 20797450 DOI: 10.1016/j.resp.2010.08.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 08/16/2010] [Accepted: 08/17/2010] [Indexed: 02/07/2023]
Abstract
In recent years, it has become evident that reactive oxygen species (ROS) play a critical role in the regulation of several physiological and pathophysiological processes. Herein we review the main sources, targets and pathophysiological roles of ROS in pulmonary vascular smooth muscle. Mitochondria and NADPH oxidases represent the major sources of ROS in vascular cells. In addition, ROS can be produced by different pathways of arachidonic acid metabolism, endothelial NO synthase (eNOS) and xantine oxidase. There is increasing evidence for the role of ROS, specially hydrogen peroxide, as signaling moieties to induce increase in intracellular calcium concentration ([Ca2+]i) and contraction in pulmonary artery smooth muscle cells (PASMC) through the modulation of a variety of targets, such as Rho kinases (ROCK), protein kinase C (PKC), voltage-gated potassium K+ (Kv) channels and ryanodine receptors (RyR). Thus, an increase in ROS has been reported to contribute to the responses induced by different vasoconstrictor stimuli, including hypoxia. Finally, results from recent studies highlighting the involvement of ROS in the development of pulmonary hypertension are discussed in the present paper.
Collapse
Affiliation(s)
- Francisco Perez-Vizcaino
- Department of Pharmacology, School of Medicine, University Complutense of Madrid and Ciber Enfermedades Respiratorias (Ciberes), 28040 Madrid, Spain.
| | | | | |
Collapse
|
47
|
Wang YX, Zheng YM. Role of ROS signaling in differential hypoxic Ca2+ and contractile responses in pulmonary and systemic vascular smooth muscle cells. Respir Physiol Neurobiol 2010; 174:192-200. [PMID: 20713188 DOI: 10.1016/j.resp.2010.08.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Revised: 08/06/2010] [Accepted: 08/09/2010] [Indexed: 01/25/2023]
Abstract
Hypoxia causes a large increase in [Ca2+]i and attendant contraction in pulmonary artery smooth muscle cells (PASMCs), but not in systemic artery SMCs. The different responses meet the respective functional needs in these two distinct vascular myocytes; however, the underlying molecular mechanisms are not well known. We and other investigators have provided extensive evidence to reveal that voltage-dependent K+ (KV) channels, canonical transient receptor potential (TRPC) channels, ryanodine receptor Ca2+ release channels (RyRs), cyclic adenosine diphosphate-ribose, FK506 binding protein 12.6, protein kinase C, NADPH oxidase and reactive oxygen species (ROS) are the essential effectors and signaling intermediates in the hypoxic increase in [Ca2+]i in PASMCs and HPV, but they may not primarily underlie the diverse cellular responses in pulmonary and systemic vascular myocytes. Hypoxia significantly increases mitochondrial ROS generation in PASMCs, which can induce intracellular Ca2+ release by opening RyRs, and may also cause extracellular Ca2+ influx by inhibiting KV channels and activating TRPC channels, leading to a large increase in [Ca2+]i in PASMCs and HPV. In contrast, hypoxia has no or a minor effect on mitochondrial ROS generation in systemic SMCs, thereby causing no change or a negligible increase in [Ca2+]i and contraction. Further preliminary work indicates that Rieske iron-sulfur protein in the mitochondrial complex III may perhaps serve as a key initial molecular determinant for the hypoxic increase in [Ca2+]i in PASMCs and HPV, suggesting its potential important role in different cellular changes to respond to hypoxic stimulation in pulmonary and systemic artery myocytes. All these findings have greatly improved our understanding of the molecular processes for the differential hypoxic Ca2+ and contractile responses in vascular SMCs from distinct pulmonary and systemic circulation systems.
Collapse
Affiliation(s)
- Yong-Xiao Wang
- Center for Cardiovascular Sciences, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA.
| | | |
Collapse
|
48
|
Niu W, Bilan PJ, Yu J, Gao J, Boguslavsky S, Schertzer JD, Chu G, Yao Z, Klip A. PKCε regulates contraction-stimulated GLUT4 traffic in skeletal muscle cells. J Cell Physiol 2010; 226:173-80. [DOI: 10.1002/jcp.22320] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
49
|
Zheng YF, Dai DZ, Dai Y. NaHS ameliorates diabetic vascular injury by correcting depressed connexin 43 and 40 in the vasculature in streptozotocin-injected rats. J Pharm Pharmacol 2010; 62:883-9. [DOI: 10.1211/jpp.62.05.0009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
50
|
Wang YX, Zheng YM. ROS-dependent signaling mechanisms for hypoxic Ca(2+) responses in pulmonary artery myocytes. Antioxid Redox Signal 2010; 12:611-23. [PMID: 19764882 PMCID: PMC2861542 DOI: 10.1089/ars.2009.2877] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Hypoxic exposure causes pulmonary vasoconstriction, which serves as a critical physiologic process that ensures regional alveolar ventilation and pulmonary perfusion in the lungs, but may become an essential pathologic factor leading to pulmonary hypertension. Although the molecular mechanisms underlying hypoxic pulmonary vasoconstriction and associated pulmonary hypertension are uncertain, increasing evidence indicates that hypoxia can result in a significant increase in intracellular reactive oxygen species concentration ([ROS](i)) through the mitochondrial electron-transport chain in pulmonary artery smooth muscle cells (PASMCs). The increased mitochondrial ROS subsequently activate protein kinase C-epsilon (PKCepsilon) and NADPH oxidase (Nox), providing positive mechanisms that further increase [ROS](i). ROS may directly cause extracellular Ca(2+) influx by inhibiting voltage-dependent K(+) (K(V)) channels and opening of store-operated Ca(2+) (SOC) channels, as well as intracellular Ca(2+) release by activating ryanodine receptors (RyRs), leading to an increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) and associated contraction. In concert with ROS, PKCepsilon may also affect K(V) channels, SOC channels, and RyRs, contributing to hypoxic Ca(2+) and contractile responses in PASMCs.
Collapse
Affiliation(s)
- Yong-Xiao Wang
- Center for Cardiovascular Sciences, Albany Medical College, New York 12208, USA.
| | | |
Collapse
|