1
|
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
|
2
|
Nakamura K, Akagi S, Ejiri K, Taya S, Saito Y, Kuroda K, Takaya Y, Toh N, Nakayama R, Katanosaka Y, Yuasa S. Pathophysiology of Group 3 Pulmonary Hypertension Associated with Lung Diseases and/or Hypoxia. Int J Mol Sci 2025; 26:835. [PMID: 39859549 PMCID: PMC11765551 DOI: 10.3390/ijms26020835] [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: 11/18/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Pulmonary hypertension associated with lung diseases and/or hypoxia is classified as group 3 in the clinical classification of pulmonary hypertension. The efficacy of existing selective pulmonary vasodilators for group 3 pulmonary hypertension is still unknown, and it is currently associated with a poor prognosis. The mechanisms by which pulmonary hypertension occurs include hypoxic pulmonary vasoconstriction, pulmonary vascular remodeling, a decrease in pulmonary vascular beds, endothelial dysfunction, endothelial-to-mesenchymal transition, mitochondrial dysfunction, oxidative stress, hypoxia-inducible factors (HIFs), inflammation, microRNA, and genetic predisposition. Among these, hypoxic pulmonary vasoconstriction and subsequent pulmonary vascular remodeling are characteristic factors involving the pulmonary vasculature and are the focus of this review. Several factors have been reported to mediate vascular remodeling induced by hypoxic pulmonary vasoconstriction, such as HIF-1α and mechanosensors, including TRP channels. New therapies that target novel molecules, such as mechanoreceptors, to inhibit vascular remodeling are awaited.
Collapse
Affiliation(s)
- Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
- Center for Advanced Heart Failure, Okayama University Hospital, Okayama 700-8558, Japan
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Kentaro Ejiri
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Satoshi Taya
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Kazuhiro Kuroda
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Yoichi Takaya
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Norihisa Toh
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Rie Nakayama
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| | - Yuki Katanosaka
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan;
- Graduate School of Pharmaceutical Sciences, Kinjo Gakuin University, Nagoya 463-8521, Japan
| | - Shinsuke Yuasa
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (S.A.); (K.E.); (S.T.); (Y.S.); (K.K.); (Y.T.); (N.T.); (R.N.); (S.Y.)
| |
Collapse
|
3
|
Xiao R, Liu J, Shi L, Zhang T, Liu J, Qiu S, Ruiz M, Dupuis J, Zhu L, Wang L, Wang Z, Hu Q. Au-modified ceria nanozyme prevents and treats hypoxia-induced pulmonary hypertension with greatly improved enzymatic activity and safety. J Nanobiotechnology 2024; 22:492. [PMID: 39160624 PMCID: PMC11331617 DOI: 10.1186/s12951-024-02738-4] [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: 11/23/2023] [Accepted: 07/24/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Despite recent advances the prognosis of pulmonary hypertension remains poor and warrants novel therapeutic options. Extensive studies, including ours, have revealed that hypoxia-induced pulmonary hypertension is associated with high oxidative stress. Cerium oxide nanozyme or nanoparticles (CeNPs) have displayed catalytic activity mimicking both catalase and superoxide dismutase functions and have been widely used as an anti-oxidative stress approach. However, whether CeNPs can attenuate hypoxia-induced pulmonary vascular oxidative stress and pulmonary hypertension is unknown. RESULTS In this study, we designed a new ceria nanozyme or nanoparticle (AuCeNPs) exhibiting enhanced enzyme activity. The AuCeNPs significantly blunted the increase of reactive oxygen species and intracellular calcium concentration while limiting proliferation of pulmonary artery smooth muscle cells and pulmonary vasoconstriction in a model of hypoxia-induced pulmonary hypertension. In addition, the inhalation of nebulized AuCeNPs, but not CeNPs, not only prevented but also blunted hypoxia-induced pulmonary hypertension in rats. The benefits of AuCeNPs were associated with limited increase of intracellular calcium concentration as well as enhancement of extracellular calcium-sensing receptor (CaSR) activity and expression in rat pulmonary artery smooth muscle cells. Nebulised AuCeNPs showed a favorable safety profile, systemic arterial pressure, liver and kidney function, plasma Ca2+ level, and blood biochemical parameters were not affected. CONCLUSION We conclude that AuCeNPs is an improved reactive oxygen species scavenger that effectively prevents and treats hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Rui Xiao
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Lin Shi
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Ting Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Jie Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Shuyi Qiu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montréal, Québec, Canada
| | - Jocelyn Dupuis
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of medicine, Université de Montréal, Montréal, Québec, Canada
| | - Liping Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China.
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China.
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan, China.
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, HUST, Wuhan, China.
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan, China.
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, HUST, Wuhan, China.
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China.
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China.
| |
Collapse
|
4
|
Ahmed ASI, Blood AB, Zhang L. Hypoxia-induced pulmonary hypertension in adults and newborns: implications for drug development. Drug Discov Today 2024; 29:104015. [PMID: 38719143 PMCID: PMC11936511 DOI: 10.1016/j.drudis.2024.104015] [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: 02/15/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
Chronic hypoxia-induced pulmonary hypertension (CHPH) presents a complex challenge, characterized by escalating pulmonary vascular resistance and remodeling, threatening both newborns and adults with right heart failure. Despite advances in understanding the pathobiology of CHPH, its molecular intricacies remain elusive, particularly because of the multifaceted nature of arterial remodeling involving the adventitia, media, and intima. Cellular imbalance arises from hypoxia-induced mitochondrial disturbances and oxidative stress, reflecting the diversity in pulmonary hypertension (PH) pathology. In this review, we highlight prominent mechanisms causing CHPH in adults and newborns, and emerging therapeutic targets of potential pharmaceuticals.
Collapse
Affiliation(s)
- Abu Shufian Ishtiaq Ahmed
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Arlin B Blood
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
5
|
Fang X, Zhang Y, Wu H, Wang H, Miao R, Wei J, Zhang Y, Tian J, Tong X. Mitochondrial regulation of diabetic endothelial dysfunction: Pathophysiological links. Int J Biochem Cell Biol 2024; 170:106569. [PMID: 38556159 DOI: 10.1016/j.biocel.2024.106569] [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: 12/07/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
Micro- and macrovascular complications frequently occur in patients with diabetes, with endothelial dysfunction playing a key role in the development and progression of the complications. For the early diagnosis and optimal treatment of vascular complications associated with diabetes, it is imperative to comprehend the cellular and molecular mechanisms governing the function of diabetic endothelial cells. Mitochondria function as crucial sensors of environmental and cellular stress regulating endothelial cell viability, structural integrity and function. Impaired mitochondrial quality control mechanisms and mitochondrial dysfunction are the main features of endothelial damage. Hence, targeted mitochondrial therapy is considered promising novel therapeutic options in vascular complications of diabetes. In this review, we focus on the mitochondrial functions in the vascular endothelial cells and the pathophysiological role of mitochondria in diabetic endothelial dysfunction, aiming to provide a reference for related drug development and clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Xinyi Fang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanjiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Haoran Wu
- Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Han Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiahua Wei
- Graduate College, Changchun University of Chinese Medicine, Jilin 130117, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Xiaolin Tong
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
6
|
Li X, Cai P, Tang X, Wu Y, Zhang Y, Rong X. Lactylation Modification in Cardiometabolic Disorders: Function and Mechanism. Metabolites 2024; 14:217. [PMID: 38668345 PMCID: PMC11052226 DOI: 10.3390/metabo14040217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Cardiovascular disease (CVD) is recognized as the primary cause of mortality and morbidity on a global scale, and developing a clear treatment is an important tool for improving it. Cardiometabolic disorder (CMD) is a syndrome resulting from the combination of cardiovascular, endocrine, pro-thrombotic, and inflammatory health hazards. Due to their complex pathological mechanisms, there is a lack of effective diagnostic and treatment methods for cardiac metabolic disorders. Lactylation is a type of post-translational modification (PTM) that plays a regulatory role in various cellular physiological processes by inducing changes in the spatial conformation of proteins. Numerous studies have reported that lactylation modification plays a crucial role in post-translational modifications and is closely related to cardiac metabolic diseases. This article discusses the molecular biology of lactylation modifications and outlines the roles and mechanisms of lactylation modifications in cardiometabolic disorders, offering valuable insights for the diagnosis and treatment of such conditions.
Collapse
Affiliation(s)
- Xu Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Pingdong Cai
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xinyuan Tang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yingzi Wu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xianglu Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
7
|
Santos EW, Khatoon S, Di Mise A, Zheng YM, Wang YX. Mitochondrial Dynamics in Pulmonary Hypertension. Biomedicines 2023; 12:53. [PMID: 38255160 PMCID: PMC10813473 DOI: 10.3390/biomedicines12010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Mitochondria are essential organelles for energy production, calcium homeostasis, redox signaling, and other cellular responses involved in pulmonary vascular biology and disease processes. Mitochondrial homeostasis depends on a balance in mitochondrial fusion and fission (dynamics). Mitochondrial dynamics are regulated by a viable circadian clock. Hypoxia and nicotine exposure can cause dysfunctions in mitochondrial dynamics, increases in mitochondrial reactive oxygen species generation and calcium concentration, and decreases in ATP production. These mitochondrial changes contribute significantly to pulmonary vascular oxidative stress, inflammatory responses, contractile dysfunction, pathologic remodeling, and eventually pulmonary hypertension. In this review article, therefore, we primarily summarize recent advances in basic, translational, and clinical studies of circadian roles in mitochondrial metabolism in the pulmonary vasculature. This knowledge may not only be crucial to fully understanding the development of pulmonary hypertension, but also greatly help to create new therapeutic strategies for treating this devastating disease and other related pulmonary disorders.
Collapse
Affiliation(s)
- Ed Wilson Santos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Subika Khatoon
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Annarita Di Mise
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona, 4, 70125 Bari, Italy
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| |
Collapse
|
8
|
Yan S, Sheak JR, Walker BR, Jernigan NL, Resta TC. Contribution of Mitochondrial Reactive Oxygen Species to Chronic Hypoxia-Induced Pulmonary Hypertension. Antioxidants (Basel) 2023; 12:2060. [PMID: 38136180 PMCID: PMC10741244 DOI: 10.3390/antiox12122060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Pulmonary hypertension (PH) resulting from chronic hypoxia (CH) occurs in patients with chronic obstructive pulmonary diseases, sleep apnea, and restrictive lung diseases, as well as in residents at high altitude. Previous studies from our group and others demonstrate a detrimental role of reactive oxygen species (ROS) in the pathogenesis of CH-induced PH, although the subcellular sources of ROS are not fully understood. We hypothesized that mitochondria-derived ROS (mtROS) contribute to enhanced vasoconstrictor reactivity and PH following CH. To test the hypothesis, we exposed rats to 4 weeks of hypobaric hypoxia (PB ≈ 380 mmHg), with control rats housed in ambient air (PB ≈ 630 mmHg). Chronic oral administration of the mitochondria-targeted antioxidant MitoQ attenuated CH-induced decreases in pulmonary artery (PA) acceleration time, increases in right ventricular systolic pressure, right ventricular hypertrophy, and pulmonary arterial remodeling. In addition, endothelium-intact PAs from CH rats exhibited a significantly greater basal tone compared to those from control animals, as was eliminated via MitoQ. CH also augmented the basal tone in endothelium-disrupted PAs, a response associated with increased mtROS production in primary PA smooth muscle cells (PASMCs) from CH rats. However, we further uncovered an effect of NO synthase inhibition with Nω-nitro-L-arginine (L-NNA) to unmask a potent endothelial vasoconstrictor influence that accentuates mtROS-dependent vasoconstriction following CH. This basal tone augmentation in the presence of L-NNA disappeared following combined endothelin A and B receptor blockade with BQ123 and BQ788. The effects of using CH to augment vasoconstriction and PASMC mtROS production in exogenous endothelin 1 (ET-1) were similarly prevented by MitoQ. We conclude that mtROS participate in the development of CH-induced PH. Furthermore, mtROS signaling in PASMCs is centrally involved in enhanced pulmonary arterial constriction following CH, a response potentiated by endogenous ET-1.
Collapse
Affiliation(s)
| | | | | | | | - Thomas C. Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA (J.R.S.); (B.R.W.); (N.L.J.)
| |
Collapse
|
9
|
Burtscher J, Hohenauer E, Burtscher M, Millet GP, Egg M. Environmental and behavioral regulation of HIF-mitochondria crosstalk. Free Radic Biol Med 2023; 206:63-73. [PMID: 37385566 DOI: 10.1016/j.freeradbiomed.2023.06.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/05/2023] [Accepted: 06/19/2023] [Indexed: 07/01/2023]
Abstract
Reduced oxygen availability (hypoxia) can lead to cell and organ damage. Therefore, aerobic species depend on efficient mechanisms to counteract detrimental consequences of hypoxia. Hypoxia inducible factors (HIFs) and mitochondria are integral components of the cellular response to hypoxia and coordinate both distinct and highly intertwined adaptations. This leads to reduced dependence on oxygen, improved oxygen supply, maintained energy provision by metabolic remodeling and tapping into alternative pathways and increased resilience to hypoxic injuries. On one hand, many pathologies are associated with hypoxia and hypoxia can drive disease progression, for example in many cancer and neurological diseases. But on the other hand, controlled induction of hypoxia responses via HIFs and mitochondria can elicit profound health benefits and increase resilience. To tackle pathological hypoxia conditions or to apply health-promoting hypoxia exposures efficiently, cellular and systemic responses to hypoxia need to be well understood. Here we first summarize the well-established link between HIFs and mitochondria in orchestrating hypoxia-induced adaptations and then outline major environmental and behavioral modulators of their interaction that remain poorly understood.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland.
| | - Erich Hohenauer
- Rehabilitation and Exercise Science Laboratory (RES Lab), Department of Business Economics, Health and Social Care, University of Applied Sciences and Arts of Southern Switzerland, Landquart, Switzerland; International University of Applied Sciences THIM, Landquart, Switzerland; Department of Neurosciences and Movement Science, University of Fribourg, Fribourg, Switzerland; Department of Movement and Sport Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland
| | - Margit Egg
- Institute of Zoology, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
10
|
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
|
11
|
Ryanto GRT, Suraya R, Nagano T. Mitochondrial Dysfunction in Pulmonary Hypertension. Antioxidants (Basel) 2023; 12:372. [PMID: 36829931 PMCID: PMC9952650 DOI: 10.3390/antiox12020372] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/21/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a multi-etiological condition with a similar hemodynamic clinical sign and end result of right heart failure. Although its causes vary, a similar link across all the classifications is the presence of mitochondrial dysfunction. Mitochondria, as the powerhouse of the cells, hold a number of vital roles in maintaining normal cellular homeostasis, including the pulmonary vascular cells. As such, any disturbance in the normal functions of mitochondria could lead to major pathological consequences. The Warburg effect has been established as a major finding in PH conditions, but other mitochondria-related metabolic and oxidative stress factors have also been reported, making important contributions to the progression of pulmonary vascular remodeling that is commonly found in PH pathophysiology. In this review, we will discuss the role of the mitochondria in maintaining a normal vasculature, how it could be altered during pulmonary vascular remodeling, and the therapeutic options available that can treat its dysfunction.
Collapse
Affiliation(s)
- Gusty Rizky Teguh Ryanto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Kobe 658-8558, Japan
| | - Ratoe Suraya
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| |
Collapse
|
12
|
Breault NM, Wu D, Dasgupta A, Chen KH, Archer SL. Acquired disorders of mitochondrial metabolism and dynamics in pulmonary arterial hypertension. Front Cell Dev Biol 2023; 11:1105565. [PMID: 36819102 PMCID: PMC9933518 DOI: 10.3389/fcell.2023.1105565] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is an orphan disease of the cardiopulmonary unit that reflects an obstructive pulmonary vasculopathy and presents with hypertrophy, inflammation, fibrosis, and ultimately failure of the right ventricle (RVF). Despite treatment using pulmonary hypertension (PH)-targeted therapies, persistent functional impairment reduces the quality of life for people with PAH and death from RVF occurs in approximately 40% of patients within 5 years of diagnosis. PH-targeted therapeutics are primarily vasodilators and none, alone or in combination, are curative. This highlights a need to therapeutically explore molecular targets in other pathways that are involved in the pathogenesis of PAH. Several candidate pathways in PAH involve acquired mitochondrial dysfunction. These mitochondrial disorders include: 1) a shift in metabolism related to increased expression of pyruvate dehydrogenase kinase and pyruvate kinase, which together increase uncoupled glycolysis (Warburg metabolism); 2) disruption of oxygen-sensing related to increased expression of hypoxia-inducible factor 1α, resulting in a state of pseudohypoxia; 3) altered mitochondrial calcium homeostasis related to impaired function of the mitochondrial calcium uniporter complex, which elevates cytosolic calcium and reduces intramitochondrial calcium; and 4) abnormal mitochondrial dynamics related to increased expression of dynamin-related protein 1 and its binding partners, such as mitochondrial dynamics proteins of 49 kDa and 51 kDa, and depressed expression of mitofusin 2, resulting in increased mitotic fission. These acquired mitochondrial abnormalities increase proliferation and impair apoptosis in most pulmonary vascular cells (including endothelial cells, smooth muscle cells and fibroblasts). In the RV, Warburg metabolism and induction of glutaminolysis impairs bioenergetics and promotes hypokinesis, hypertrophy, and fibrosis. This review will explore our current knowledge of the causes and consequences of disordered mitochondrial function in PAH.
Collapse
Affiliation(s)
- Nolan M. Breault
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Danchen Wu
- Department of Medicine, Queen’s University, Kingston, ON, Canada,*Correspondence: Danchen Wu, ; Stephen L. Archer,
| | - Asish Dasgupta
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Kuang-Hueih Chen
- Department of Medicine, Queen’s University, Kingston, ON, Canada
| | - Stephen L. Archer
- Department of Medicine, Queen’s University, Kingston, ON, Canada,Queen’s Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Department of Medicine, Queen’s University, Kingston, ON, Canada,*Correspondence: Danchen Wu, ; Stephen L. Archer,
| |
Collapse
|
13
|
DeVallance ER, Dustin CM, de Jesus DS, Ghouleh IA, Sembrat JC, Cifuentes-Pagano E, Pagano PJ. Specificity Protein 1-Mediated Promotion of CXCL12 Advances Endothelial Cell Metabolism and Proliferation in Pulmonary Hypertension. Antioxidants (Basel) 2022; 12:71. [PMID: 36670936 PMCID: PMC9854820 DOI: 10.3390/antiox12010071] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/31/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare yet devastating and incurable disease with few treatment options. The underlying mechanisms of PAH appear to involve substantial cellular proliferation and vascular remodeling, causing right ventricular overload and eventual heart failure. Recent evidence suggests a significant seminal role of the pulmonary endothelium in the initiation and promotion of PAH. Our previous work identified elevated reactive oxygen species (ROS)-producing enzyme NADPH oxidase 1 (NOX1) in human pulmonary artery endothelial cells (HPAECs) of PAH patients promoting endothelial cell proliferation in vitro. In this study, we interrogated chemokine CXCL12's (aka SDF-1) role in EC proliferation under the control of NOX1 and specificity protein 1 (Sp1). We report here that NOX1 can drive hypoxia-induced endothelial CXCL12 expression via the transcription factor Sp1 leading to HPAEC proliferation and migration. Indeed, NOX1 drove hypoxia-induced Sp1 activation, along with an increased capacity of Sp1 to bind cognate promoter regions in the CXCL12 promoter. Sp1 activation induced elevated expression of CXCL12 in hypoxic HPAECs, supporting downstream induction of expression at the CXCL12 promoter via NOX1 activity. Pathological levels of CXCL12 mimicking those reported in human PAH patient serum restored EC proliferation impeded by specific NOX1 inhibitor. The translational relevance of our findings is highlighted by elevated NOX1 activity, Sp1 activation, and CXCL12 expression in explanted lung samples from PAH patients compared to non-PAH controls. Analysis of phosphofructokinase, glucose-6-phosphate dehydrogenase, and glutaminase activity revealed that CXCL12 induces glutamine and glucose metabolism, which are foundational to EC cell proliferation. Indeed, in explanted human PAH lungs, demonstrably higher glutaminase activity was detected compared to healthy controls. Finally, infusion of recombinant CXCL12 into healthy mice amplified pulmonary arterial pressure, right ventricle remodeling, and elevated glucose and glutamine metabolism. Together these data suggest a central role for a novel NOX1-Sp1-CXCL12 pathway in mediating PAH phenotype in the lung endothelium.
Collapse
Affiliation(s)
- Evan R. DeVallance
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Christopher M. Dustin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daniel Simoes de Jesus
- William Harvey Research Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Imad Al Ghouleh
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Cardiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - John C. Sembrat
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Eugenia Cifuentes-Pagano
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Patrick J. Pagano
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
14
|
Mohammadi A, Higazy R, Gauda EB. PGC-1α activity and mitochondrial dysfunction in preterm infants. Front Physiol 2022; 13:997619. [PMID: 36225305 PMCID: PMC9548560 DOI: 10.3389/fphys.2022.997619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Extremely low gestational age neonates (ELGANs) are born in a relatively hyperoxic environment with weak antioxidant defenses, placing them at high risk for mitochondrial dysfunction affecting multiple organ systems including the nervous, respiratory, ocular, and gastrointestinal systems. The brain and lungs are highly affected by mitochondrial dysfunction and dysregulation in the neonate, causing white matter injury (WMI) and bronchopulmonary dysplasia (BPD), respectively. Adequate mitochondrial function is important in providing sufficient energy for organ development as it relates to alveolarization and axonal myelination and decreasing oxidative stress via reactive oxygen species (ROS) and reactive nitrogen species (RNS) detoxification. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) is a master regulator of mitochondrial biogenesis and function. Since mitochondrial dysfunction is at the root of WMI and BPD pathobiology, exploring therapies that can regulate PGC-1α activity may be beneficial. This review article describes several promising therapeutic agents that can mitigate mitochondrial dysfunction through direct and indirect activation and upregulation of the PGC-1α pathway. Metformin, resveratrol, omega 3 fatty acids, montelukast, L-citrulline, and adiponectin are promising candidates that require further pre-clinical and clinical studies to understand their efficacy in decreasing the burden of disease from WMI and BPD in preterm infants.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Randa Higazy
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
| | - Estelle B. Gauda
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Mitochondrial Regulation of the Hypoxia-Inducible Factor in the Development of Pulmonary Hypertension. J Clin Med 2022; 11:jcm11175219. [PMID: 36079149 PMCID: PMC9457092 DOI: 10.3390/jcm11175219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary hypertension (PH) is a severe progressive lung disorder characterized by pulmonary vasoconstriction and vascular remodeling, culminating in right-sided heart failure and increased mortality. Data from animal models and human subjects demonstrated that hypoxia-inducible factor (HIF)-related signaling is essential in the progression of PH. This review summarizes the regulatory pathways and mechanisms of HIF-mediated signaling, emphasizing the role of mitochondria in HIF regulation and PH pathogenesis. We also try to determine the potential to therapeutically target the components of the HIF system for the management of PH.
Collapse
|
16
|
Escada-Rebelo S, Cristo MI, Ramalho-Santos J, Amaral S. Mitochondria-Targeted Compounds to Assess and Improve Human Sperm Function. Antioxid Redox Signal 2022; 37:451-480. [PMID: 34847742 DOI: 10.1089/ars.2021.0238] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Currently 10%-15% of couples in reproductive age face infertility issues. More importantly, male factor contributes to 50% of these cases (either alone or in combination with female causes). Among various reasons, impaired sperm function is the main cause for male infertility. Furthermore, mitochondrial dysfunction and oxidative stress due to increased reactive oxygen species (ROS) production, particularly of mitochondrial origin, are believed to be the main contributors. Recent Advances: Mitochondrial dysfunction, particularly due to increased ROS production, has often been linked to impaired sperm function/quality. For decades, different methods and approaches have been developed to assess mitochondrial features that might correlate with sperm functionality. This connection is now completely accepted, with mitochondrial functionality assessment used more commonly as a readout of sperm functionality. More recently, mitochondria-targeted compounds are on the frontline for both assessment and therapeutic approaches. Critical Issues: In this review, we summarize the current methods for assessing key mitochondrial parameters known to reflect sperm quality as well as therapeutic strategies using mitochondria-targeted antioxidants aiming to improve sperm function in various situations, particularly after sperm cryopreservation. Future Directions: Although more systematic research is needed, mitochondria-targeted compounds definitely represent a promising tool to assess as well as to protect and improve sperm function. Antioxid. Redox Signal. 37, 451-480.
Collapse
Affiliation(s)
- Sara Escada-Rebelo
- PhD Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Biology of Reproduction and Stem Cell Group, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,IIIUC - Institute for Interdisciplinary Research, Casa Costa Alemão, University of Coimbra, Coimbra, Portugal
| | - Maria Inês Cristo
- Biology of Reproduction and Stem Cell Group, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - João Ramalho-Santos
- Biology of Reproduction and Stem Cell Group, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Sandra Amaral
- Biology of Reproduction and Stem Cell Group, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,IIIUC - Institute for Interdisciplinary Research, Casa Costa Alemão, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
17
|
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
|
18
|
Wang R, Pan J, Han J, Gong M, Liu L, Zhang Y, Liu Y, Wang D, Tang Q, Wu N, Wang L, Yan J, Li H, Yuan Y. Melatonin Attenuates Dasatinib-Aggravated Hypoxic Pulmonary Hypertension via Inhibiting Pulmonary Vascular Remodeling. Front Cardiovasc Med 2022; 9:790921. [PMID: 35402542 PMCID: PMC8987569 DOI: 10.3389/fcvm.2022.790921] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Dasatinib treatment is approved as first-line therapy for chronic myeloid leukemia. However, pulmonary hypertension (PH) is a highly morbid and often fatal side-effect of dasatinib, characterized by progressive pulmonary vascular remodeling. Melatonin exerts strong antioxidant capacity against the progression of cardiovascular system diseases. The present work aimed to investigate the effect of melatonin on dasatinib-aggravated hypoxic PH and explore its possible mechanisms. Dasatinib-aggravated rat experimental model of hypoxic PH was established by utilizing dasatinib under hypoxia. The results indicated that melatonin could attenuate dasatinib-aggravated pulmonary pressure and vascular remodeling in rats under hypoxia. Additionally, melatonin attenuated the activity of XO, the content of MDA, the expression of NOX4, and elevated the activity of CAT, GPx, and SOD, the expression of SOD2, which were caused by dasatinib under hypoxia. In vitro, dasatinib led to decreased LDH activity and production of NO in human pulmonary microvascular endothelial cells (HPMECs), moreover increased generation of ROS, and expression of NOX4 both in HPMECs and primary rat pulmonary arterial smooth muscle cells (PASMCs) under hypoxia. Dasatinib up-regulated the expression of cleaved caspase-3 and the ratio of apoptotic cells in HPMECs, and also elevated the percentage of S phase and the expression of Cyclin D1 in primary PASMCs under hypoxia. Melatonin ameliorated dasatinib-aggravated oxidative damage and apoptosis in HPMECs, meanwhile reduced oxidative stress level, proliferation, and repressed the stability of HIF1-α protein in PASMCs under hypoxia. In conclusion, melatonin significantly attenuates dasatinib-aggravated hypoxic PH by inhibiting pulmonary vascular remodeling in rats. The possible mechanisms involved protecting endothelial cells and inhibiting abnormal proliferation of smooth muscle cells. Our findings may suggest that melatonin has potential clinical value as a therapeutic approach to alleviate dasatinib-aggravated hypoxic PH.
Collapse
Affiliation(s)
- Rui Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
| | - Jinjin Pan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jinzhen Han
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Miaomiao Gong
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Liang Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yunlong Zhang
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Ying Liu
- The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Dingyou Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Qing Tang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Na Wu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Lin Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jinsong Yan
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Second Hospital of Dalian Medical University, Dalian, China
- Jinsong Yan,
| | - Hua Li
- College of Pharmacy, Dalian Medical University, Dalian, China
- Hua Li,
| | - Yuhui Yuan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- *Correspondence: Yuhui Yuan,
| |
Collapse
|
19
|
Madhu NR, Sarkar B, Slama P, Jha NK, Ghorai SK, Jana SK, Govindasamy K, Massanyi P, Lukac N, Kumar D, Kalita JC, Kesari KK, Roychoudhury S. Effect of Environmental Stressors, Xenobiotics, and Oxidative Stress on Male Reproductive and Sexual Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1391:33-58. [PMID: 36472815 DOI: 10.1007/978-3-031-12966-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This article examines the environmental factor-induced oxidative stress (OS) and their effects on male reproductive and sexual health. There are several factors that induce OS, i.e. radition, metal contamination, xenobiotic compounds, and cigarette smoke and lead to cause toxicity in the cells through metabolic or bioenergetic processes. These environmental factors may produce free radicals and enhance the reactive oxygen species (ROS). Free radicals are molecules that include oxygen and disbalance the amount of electrons that can create major chemical chains in the body and cause oxidation. Oxidative damage to cells may impair male fertility and lead to abnormal embryonic development. Moreover, it does not only cause a vast number of health issues such as ageing, cancer, atherosclerosis, insulin resistance, diabetes mellitus, cardiovascular diseases, ischemia-reperfusion injury, and neurodegenerative disorders but also decreases the motility of spermatozoa while increasing sperm DNA damage, impairing sperm mitochondrial membrane lipids and protein kinases. This chapter mainly focuses on the environmental stressors with further discussion on the mechanisms causing congenital impairments due to poor sexual health and transmitting altered signal transduction pathways in male gonadal tissues.
Collapse
Affiliation(s)
- Nithar Ranjan Madhu
- Department of Zoology, Acharya Prafulla Chandra College, New Barrackpore, Kolkata, West Bengal, India
| | - Bhanumati Sarkar
- Department of Botany, Acharya Prafulla Chandra College, New Barrackpore, Kolkata, West Bengal, India
| | - Petr Slama
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, India
| | | | - Sandip Kumar Jana
- Department of Zoology, Bajkul Milani Mahavidyalaya, Purba Medinipur, West Bengal, India
| | - Kadirvel Govindasamy
- Animal Production Division, ICAR Research Complex for NEH Region, Indian Council of Agricultural Research, Umiam, Meghalaya, India
| | - Peter Massanyi
- Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovak Republic
| | - Norbert Lukac
- Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovak Republic
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand, India
| | - Jogen C Kalita
- Department of Zoology, Gauhati University, Guwahati, India
| | | | | |
Collapse
|
20
|
He YY, Xie XM, Zhang HD, Ye J, Gencer S, van der Vorst EPC, Döring Y, Weber C, Pang XB, Jing ZC, Yan Y, Han ZY. Identification of Hypoxia Induced Metabolism Associated Genes in Pulmonary Hypertension. Front Pharmacol 2021; 12:753727. [PMID: 34803695 PMCID: PMC8602807 DOI: 10.3389/fphar.2021.753727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/11/2021] [Indexed: 01/10/2023] Open
Abstract
Objective: Pulmonary hypertension (PH) associated with hypoxia and lung disease (Group 3) is the second most common form of PH and associated with increased morbidity and mortality. This study was aimed to identify hypoxia induced metabolism associated genes (MAGs) for better understanding of hypoxic PH. Methods: Rat pulmonary arterial smooth muscle cells (PASMCs) were isolated and cultured in normoxic or hypoxic condition for 24 h. Cells were harvested for liquid chromatography-mass spectrometry analysis. Functional annotation of distinguishing metabolites was performed using Metaboanalyst. Top 10 enriched metabolite sets were selected for the identification of metabolism associated genes (MAGs) with a relevance score >8 in Genecards. Transcriptomic data from lungs of hypoxic PH in mice/rats or of PH patients were accessed from Gene Expression Omnibus (GEO) database or open-access online platform. Connectivity Map analysis was performed to identify potential compounds to reverse the metabolism associated gene profile under hypoxia stress. The construction and module analysis of the protein-protein interaction (PPI) network was performed. Hub genes were then identified and used to generate LASSO model to determine its accuracy to predict occurrence of PH. Results: A total of 36 altered metabolites and 1,259 unique MAGs were identified in rat PASMCs under hypoxia. 38 differentially expressed MAGs in mouse lungs of hypoxic PH were revealed, with enrichment in multi-pathways including regulation of glucose metabolic process, which might be reversed by drugs such as blebbistatin. 5 differentially expressed MAGs were displayed in SMCs of Sugen 5416/hypoxia induced PH rats at the single cell resolution. Furthermore, 6 hub genes (Cat, Ephx1, Gpx3, Gstm4, Gstm5, and Gsto1) out of 42 unique hypoxia induced MAGs were identified. Higher Cat, Ephx1 and lower Gsto1 were displayed in mouse lungs under hypoxia (all p < 0.05), in consistent with the alteration in lungs of PH patients. The hub gene-based LASSO model can predict the occurrence of PH (AUC = 0.90). Conclusion: Our findings revealed six hypoxia-induced metabolism associated hub genes, and shed some light on the molecular mechanism and therapeutic targets in hypoxic PH.
Collapse
Affiliation(s)
- Yang-Yang He
- School of Pharmacy, Henan University, Kaifeng, China
| | - Xin-Mei Xie
- School of Pharmacy, Henan University, Kaifeng, China
| | - Hong-Da Zhang
- State Key Laboratory of Cardiovascular Disease and FuWai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jue Ye
- State Key Laboratory of Cardiovascular Disease and FuWai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Selin Gencer
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany.,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Xiao-Bin Pang
- School of Pharmacy, Henan University, Kaifeng, China
| | - Zhi-Cheng Jing
- State Key Laboratory of Complex, Severe, and Rare Diseases, Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Yan
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Zhi-Yan Han
- State Key Laboratory of Cardiovascular Disease and FuWai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Siques P, Pena E, Brito J, El Alam S. Oxidative Stress, Kinase Activation, and Inflammatory Pathways Involved in Effects on Smooth Muscle Cells During Pulmonary Artery Hypertension Under Hypobaric Hypoxia Exposure. Front Physiol 2021; 12:690341. [PMID: 34434114 PMCID: PMC8381601 DOI: 10.3389/fphys.2021.690341] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
High-altitude exposure results in hypobaric hypoxia, which affects organisms by activating several mechanisms at the physiological, cellular, and molecular levels and triggering the development of several pathologies. One such pathology is high-altitude pulmonary hypertension (HAPH), which is initiated through hypoxic pulmonary vasoconstriction to distribute blood to more adequately ventilated areas of the lungs. Importantly, all layers of the pulmonary artery (adventitia, smooth muscle, and endothelium) contribute to or are involved in the development of HAPH. However, the principal action sites of HAPH are pulmonary artery smooth muscle cells (PASMCs), which interact with several extracellular and intracellular molecules and participate in mechanisms leading to proliferation, apoptosis, and fibrosis. This review summarizes the alterations in molecular pathways related to oxidative stress, inflammation, kinase activation, and other processes that occur in PASMCs during pulmonary hypertension under hypobaric hypoxia and proposes updates to pharmacological treatments to mitigate the pathological changes in PASMCs under such conditions. In general, PASMCs exposed to hypobaric hypoxia undergo oxidative stress mediated by Nox4, inflammation mediated by increases in interleukin-6 levels and inflammatory cell infiltration, and activation of the protein kinase ERK1/2, which lead to the proliferation of PASMCs and contribute to the development of hypobaric hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Patricia Siques
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Eduardo Pena
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Julio Brito
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Samia El Alam
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| |
Collapse
|
22
|
Escada-Rebelo S, Mora FG, Sousa AP, Almeida-Santos T, Paiva A, Ramalho-Santos J. Fluorescent probes for the detection of reactive oxygen species in human spermatozoa. Asian J Androl 2021; 22:465-471. [PMID: 31939350 PMCID: PMC7523605 DOI: 10.4103/aja.aja_132_19] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) production is a by-product of mitochondrial activity and is necessary for the acquisition of the capacitated state, a requirement for functional spermatozoa. However, an increase in oxidative stress, due to an abnormal production of ROS, has been shown to be related to loss of sperm function, highlighting the importance of an accurate detection of sperm ROS, given the specific nature of this cell. In this work, we tested a variety of commercially available fluorescent probes to detect ROS and reactive nitrogen species (RNS) in human sperm, to define their specificity. Using both flow cytometry (FC) and fluorescence microscopy (FM), we confirmed that MitoSOX™ Red and dihydroethidium (DHE) detect superoxide anion (as determined using antimycin A as a positive control), while DAF-2A detects reactive nitrogen species (namely, nitric oxide). For the first time, we also report that RedoxSensor™ Red CC-1, CellROX® Orange Reagent, and MitoPY1 seem to be mostly sensitive to hydrogen peroxide, but not superoxide. Furthermore, mean fluorescence intensity (and not percentage of labeled cells) is the main parameter that can be reproducibly monitored using this type of methodology.
Collapse
Affiliation(s)
- Sara Escada-Rebelo
- PhD Programme in Experimental Biology and Biomedicine, CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Biology of Reproduction and Stem Cell Group, CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,IIIUC - Institute for Interdisciplinary Research, Casa Costa Alemão, University of Coimbra, Coimbra 3030-789, Portugal
| | - Francisca G Mora
- Biology of Reproduction and Stem Cell Group, CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Ana P Sousa
- Biology of Reproduction and Stem Cell Group, CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Reproductive Medicine Unit, University Hospitals of Coimbra, Coimbra 3004-561, Portugal
| | - Teresa Almeida-Santos
- Biology of Reproduction and Stem Cell Group, CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Reproductive Medicine Unit, University Hospitals of Coimbra, Coimbra 3004-561, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra 3000-370, Portugal
| | - Artur Paiva
- Clinical Pathology Unit, University Hospitals of Coimbra, Coimbra 3004-561, Portugal
| | - João Ramalho-Santos
- Biology of Reproduction and Stem Cell Group, CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-504, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra 3000-456, Portugal
| |
Collapse
|
23
|
Hypoxia and the integrated stress response promote pulmonary hypertension and preeclampsia: Implications in drug development. Drug Discov Today 2021; 26:2754-2773. [PMID: 34302972 DOI: 10.1016/j.drudis.2021.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/31/2021] [Accepted: 07/14/2021] [Indexed: 11/21/2022]
Abstract
Chronic hypoxia is a common cause of pulmonary hypertension, preeclampsia, and intrauterine growth restriction (IUGR). The molecular mechanisms underlying these diseases are not completely understood. Chronic hypoxia may induce the generation of reactive oxygen species (ROS) in mitochondria, promote endoplasmic reticulum (ER) stress, and result in the integrated stress response (ISR) in the pulmonary artery and uteroplacental tissues. Numerous studies have implicated hypoxia-inducible factors (HIFs), oxidative stress, and ER stress/unfolded protein response (UPR) in the development of pulmonary hypertension, preeclampsia and IUGR. This review highlights the roles of HIFs, mitochondria-derived ROS and UPR, as well as their interplay, in the pathogenesis of pulmonary hypertension and preeclampsia, and their implications in drug development.
Collapse
|
24
|
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
|
25
|
Brand MD. Riding the tiger - physiological and pathological effects of superoxide and hydrogen peroxide generated in the mitochondrial matrix. Crit Rev Biochem Mol Biol 2020; 55:592-661. [PMID: 33148057 DOI: 10.1080/10409238.2020.1828258] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Elevated mitochondrial matrix superoxide and/or hydrogen peroxide concentrations drive a wide range of physiological responses and pathologies. Concentrations of superoxide and hydrogen peroxide in the mitochondrial matrix are set mainly by rates of production, the activities of superoxide dismutase-2 (SOD2) and peroxiredoxin-3 (PRDX3), and by diffusion of hydrogen peroxide to the cytosol. These considerations can be used to generate criteria for assessing whether changes in matrix superoxide or hydrogen peroxide are both necessary and sufficient to drive redox signaling and pathology: is a phenotype affected by suppressing superoxide and hydrogen peroxide production; by manipulating the levels of SOD2, PRDX3 or mitochondria-targeted catalase; and by adding mitochondria-targeted SOD/catalase mimetics or mitochondria-targeted antioxidants? Is the pathology associated with variants in SOD2 and PRDX3 genes? Filtering the large literature on mitochondrial redox signaling using these criteria highlights considerable evidence that mitochondrial superoxide and hydrogen peroxide drive physiological responses involved in cellular stress management, including apoptosis, autophagy, propagation of endoplasmic reticulum stress, cellular senescence, HIF1α signaling, and immune responses. They also affect cell proliferation, migration, differentiation, and the cell cycle. Filtering the huge literature on pathologies highlights strong experimental evidence that 30-40 pathologies may be driven by mitochondrial matrix superoxide or hydrogen peroxide. These can be grouped into overlapping and interacting categories: metabolic, cardiovascular, inflammatory, and neurological diseases; cancer; ischemia/reperfusion injury; aging and its diseases; external insults, and genetic diseases. Understanding the involvement of mitochondrial matrix superoxide and hydrogen peroxide concentrations in these diseases can facilitate the rational development of appropriate therapies.
Collapse
|
26
|
Yan S, Resta TC, Jernigan NL. Vasoconstrictor Mechanisms in Chronic Hypoxia-Induced Pulmonary Hypertension: Role of Oxidant Signaling. Antioxidants (Basel) 2020; 9:E999. [PMID: 33076504 PMCID: PMC7602539 DOI: 10.3390/antiox9100999] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
Elevated resistance of pulmonary circulation after chronic hypoxia exposure leads to pulmonary hypertension. Contributing to this pathological process is enhanced pulmonary vasoconstriction through both calcium-dependent and calcium sensitization mechanisms. Reactive oxygen species (ROS), as a result of increased enzymatic production and/or decreased scavenging, participate in augmentation of pulmonary arterial constriction by potentiating calcium influx as well as activation of myofilament sensitization, therefore mediating the development of pulmonary hypertension. Here, we review the effects of chronic hypoxia on sources of ROS within the pulmonary vasculature including NADPH oxidases, mitochondria, uncoupled endothelial nitric oxide synthase, xanthine oxidase, monoamine oxidases and dysfunctional superoxide dismutases. We also summarize the ROS-induced functional alterations of various Ca2+ and K+ channels involved in regulating Ca2+ influx, and of Rho kinase that is responsible for myofilament Ca2+ sensitivity. A variety of antioxidants have been shown to have beneficial therapeutic effects in animal models of pulmonary hypertension, supporting the role of ROS in the development of pulmonary hypertension. A better understanding of the mechanisms by which ROS enhance vasoconstriction will be useful in evaluating the efficacy of antioxidants for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
| | | | - Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.Y.); (T.C.R.)
| |
Collapse
|
27
|
Han W, Fessel JP, Sherrill T, Kocurek EG, Yull FE, Blackwell TS. Enhanced Expression of Catalase in Mitochondria Modulates NF-κB-Dependent Lung Inflammation through Alteration of Metabolic Activity in Macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:1125-1134. [PMID: 32601098 PMCID: PMC7415609 DOI: 10.4049/jimmunol.1900820] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 06/08/2020] [Indexed: 01/13/2023]
Abstract
NF-κB is a reduction-oxidation-sensitive transcription factor that plays a key role in regulating the immune response. In these studies, we intended to investigate the role of mitochondrial-derived reactive oxygen species in regulating NF-κB activation by studying transgenic mice that overexpress mitochondrial-targeted human catalase (mCAT). We treated wild-type (WT) and mCAT mice with intratracheal instillation of Escherichia coli LPS and found that mCAT mice had exaggerated NF-κB activation in the lungs, increased neutrophilic alveolitis, and greater lung inflammation/injury compared with WT mice. Additional studies using bone marrow chimeras revealed that this hyperinflammatory phenotype was mediated by immune/inflammatory cells. Mechanistic studies using bone marrow-derived macrophages (BMDMs) showed that LPS treatment induced a sustained increase in NF-κB activation and expression of NF-κB-dependent inflammatory mediators in mCAT BMDMs compared with WT BMDMs. Further investigations showed that cytoplasmic, but not mitochondrial, hydrogen peroxide levels were reduced in LPS-treated mCAT BMDMs. However, mCAT macrophages exhibited increased glycolytic and oxidative metabolism, coupled with increased ATP production and an increased intracellular NADH/NAD+ ratio compared with BMDMs from WT mice. Treatment of BMDMs with lactate increased the intracellular NADH/NAD+ ratio and upregulated NF-κB activation after LPS treatment, whereas treatment with a potent inhibitor of the mitochondrial pyruvate carrier (UK5099) decreased the NADH/NAD+ ratio and reduced NF-κB activation. Taken together, these findings point to an increased availability of reducing equivalents in the form of NADH as an important mechanism by which metabolic activity modulates inflammatory signaling through the NF-κB pathway.
Collapse
Affiliation(s)
- Wei Han
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Joshua P Fessel
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Taylor Sherrill
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Emily G Kocurek
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Fiona E Yull
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232;
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232; and
- Department of Veterans Affairs Medical Center, Nashville, TN 37212
| |
Collapse
|
28
|
Xu N, Jiang S, Persson PB, Persson EAG, Lai EY, Patzak A. Reactive oxygen species in renal vascular function. Acta Physiol (Oxf) 2020; 229:e13477. [PMID: 32311827 DOI: 10.1111/apha.13477] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/22/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are produced by the aerobic metabolism. The imbalance between production of ROS and antioxidant defence in any cell compartment is associated with cell damage and may play an important role in the pathogenesis of renal disease. NADPH oxidase (NOX) family is the major ROS source in the vasculature and modulates renal perfusion. Upregulation of Ang II and adenosine activates NOX via AT1R and A1R in renal microvessels, leading to superoxide production. Oxidative stress in the kidney prompts renal vascular remodelling and increases preglomerular resistance. These are key elements in hypertension, acute and chronic kidney injury, as well as diabetic nephropathy. Renal afferent arterioles (Af), the primary resistance vessel in the kidney, fine tune renal hemodynamics and impact on blood pressure. Vice versa, ROS increase hypertension and diabetes, resulting in upregulation of Af vasoconstriction, enhancement of myogenic responses and change of tubuloglomerular feedback (TGF), which further promotes hypertension and diabetic nephropathy. In the following, we highlight oxidative stress in the function and dysfunction of renal hemodynamics. The renal microcirculatory alterations brought about by ROS importantly contribute to the pathophysiology of kidney injury, hypertension and diabetes.
Collapse
Affiliation(s)
- Nan Xu
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Shan Jiang
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
| | - Pontus B. Persson
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | | | - En Yin Lai
- Department of Physiology Zhejiang University School of Medicine Hangzhou China
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| | - Andreas Patzak
- Charité ‐ Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin Humboldt‐Universität zu Berlin, and Berlin Institute of Health Institute of Vegetative Physiology Berlin Germany
| |
Collapse
|
29
|
Niihori M, Eccles CA, Kurdyukov S, Zemskova M, Varghese MV, Stepanova AA, Galkin A, Rafikov R, Rafikova O. Rats with a Human Mutation of NFU1 Develop Pulmonary Hypertension. Am J Respir Cell Mol Biol 2020; 62:231-242. [PMID: 31461310 DOI: 10.1165/rcmb.2019-0065oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
NFU1 is a mitochondrial protein that is involved in the biosynthesis of iron-sulfur clusters, and its genetic modification is associated with disorders of mitochondrial energy metabolism. Patients with autosomal-recessive inheritance of the NFU1 mutation G208C have reduced activity of the respiratory chain Complex II and decreased levels of lipoic-acid-dependent enzymes, and develop pulmonary arterial hypertension (PAH) in ∼70% of cases. We investigated whether rats with a human mutation in NFU1 are also predisposed to PAH development. A point mutation in rat NFU1G206C (human G208C) was introduced through CRISPR/Cas9 genome editing. Hemodynamic data, tissue samples, and fresh mitochondria were collected and analyzed. NFU1G206C rats showed increased right ventricular pressure, right ventricular hypertrophy, and high levels of pulmonary artery remodeling. Computed tomography and angiography of the pulmonary vasculature indicated severe angioobliterative changes in NFU1G206C rats. Importantly, the penetrance of the PAH phenotype was found to be more prevalent in females than in males, replicating the established sex difference among patients with PAH. Male and female homozygote rats exhibited decreased expression and activity of mitochondrial Complex II, and markedly decreased pyruvate dehydrogenase activity and lipoate binding. The limited development of PAH in males correlated with the preserved levels of oligomeric NFU1, increased expression of ISCU (an alternative branch of the iron-sulfur assembly system), and increased complex IV activity. Thus, the male sex has additional plasticity to overcome the iron-sulfur cluster deficiency. Our work describes a novel, humanized rat model of NFU1 deficiency that showed mitochondrial dysfunction similar to that observed in patients and developed PAH with the same sex dimorphism.
Collapse
Affiliation(s)
- Maki Niihori
- Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona; and
| | - Cody A Eccles
- Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona; and
| | - Sergey Kurdyukov
- Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona; and
| | - Marina Zemskova
- Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona; and
| | | | - Anna A Stepanova
- Division of Neonatology, Department of Pediatrics, Columbia University, New York, New York
| | - Alexander Galkin
- Division of Neonatology, Department of Pediatrics, Columbia University, New York, New York
| | - Ruslan Rafikov
- Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona; and
| | - Olga Rafikova
- Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona; and
| |
Collapse
|
30
|
Sommer N, Alebrahimdehkordi N, Pak O, Knoepp F, Strielkov I, Scheibe S, Dufour E, Andjelković A, Sydykov A, Saraji A, Petrovic A, Quanz K, Hecker M, Kumar M, Wahl J, Kraut S, Seeger W, Schermuly RT, Ghofrani HA, Ramser K, Braun T, Jacobs HT, Weissmann N, Szibor M. Bypassing mitochondrial complex III using alternative oxidase inhibits acute pulmonary oxygen sensing. SCIENCE ADVANCES 2020; 6:eaba0694. [PMID: 32426457 PMCID: PMC7159913 DOI: 10.1126/sciadv.aba0694] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/22/2020] [Indexed: 05/02/2023]
Abstract
Mitochondria play an important role in sensing both acute and chronic hypoxia in the pulmonary vasculature, but their primary oxygen-sensing mechanism and contribution to stabilization of the hypoxia-inducible factor (HIF) remains elusive. Alteration of the mitochondrial electron flux and increased superoxide release from complex III has been proposed as an essential trigger for hypoxic pulmonary vasoconstriction (HPV). We used mice expressing a tunicate alternative oxidase, AOX, which maintains electron flux when respiratory complexes III and/or IV are inhibited. Respiratory restoration by AOX prevented acute HPV and hypoxic responses of pulmonary arterial smooth muscle cells (PASMC), acute hypoxia-induced redox changes of NADH and cytochrome c, and superoxide production. In contrast, AOX did not affect the development of chronic hypoxia-induced pulmonary hypertension and HIF-1α stabilization. These results indicate that distal inhibition of the mitochondrial electron transport chain in PASMC is an essential initial step for acute but not chronic oxygen sensing.
Collapse
Affiliation(s)
- Natascha Sommer
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Nasim Alebrahimdehkordi
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Oleg Pak
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Fenja Knoepp
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Ievgen Strielkov
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Susan Scheibe
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Eric Dufour
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | - Ana Andjelković
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | - Akylbek Sydykov
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Alireza Saraji
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Aleksandar Petrovic
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Karin Quanz
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Matthias Hecker
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Manish Kumar
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Joel Wahl
- Department of Engineering Sciences and Mathematics, Luleå University of Technology, SE-97187 Luleå, Sweden
| | - Simone Kraut
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
| | - Hossein A. Ghofrani
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
- Department of Medicine, Imperial College London, Du Cane Road, Hammersmith Campus, London W12 0NN, UK
| | - Kerstin Ramser
- Department of Engineering Sciences and Mathematics, Luleå University of Technology, SE-97187 Luleå, Sweden
| | - Thomas Braun
- Department I, Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, D-61231 Bad Nauheim, Germany
| | - Howard T. Jacobs
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, D-35392 Giessen, Germany
- Corresponding author. (M.S.); (N.W.)
| | - Marten Szibor
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
- Corresponding author. (M.S.); (N.W.)
| |
Collapse
|
31
|
Identification of key HIF-1α target genes that regulate adaptation to hypoxic conditions in Tibetan chicken embryos. Gene 2020; 729:144321. [DOI: 10.1016/j.gene.2019.144321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/10/2019] [Accepted: 12/21/2019] [Indexed: 12/11/2022]
|
32
|
Antioxidant-Conjugated Peptide Attenuated Metabolic Reprogramming in Pulmonary Hypertension. Antioxidants (Basel) 2020; 9:antiox9020104. [PMID: 31991719 PMCID: PMC7071131 DOI: 10.3390/antiox9020104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 01/11/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic cardiopulmonary disorder instigated by pulmonary vascular cell proliferation. Activation of Akt was previously reported to promote vascular remodeling. Also, the irreversible nitration of Y350 residue in Akt results in its activation. NitroAkt was increased in PAH patients and the SU5416/Hypoxia (SU/Hx) PAH model. This study investigated whether the prevention of Akt nitration in PAH by Akt targeted nitroxide-conjugated peptide (NP) could reverse vascular remodeling and metabolic reprogramming. Treatment of the SU/Hx model with NP significantly decreased nitration of Akt in lungs, attenuated right ventricle (RV) hypertrophy, and reduced RV systolic pressure. In the PAH model, Akt-nitration induces glycolysis by activation of the glucose transporter Glut4 and lactate dehydrogenase-A (LDHA). Decreased G6PD and increased GSK3β in SU/Hx additionally shunted intracellular glucose via glycolysis. The increased glycolytic rate upregulated anaplerosis due to activation of pyruvate carboxylase in a nitroAkt-dependent manner. NP treatment resolved glycolytic switch and activated collateral pentose phosphate and glycogenesis pathways. Prevention of Akt-nitration significantly controlled pyruvate in oxidative phosphorylation by decreasing lactate and increasing pyruvate dehydrogenases activities. Histopathological studies showed significantly reduced pulmonary vascular proliferation. Based on our current observation, preventing Akt-nitration by using an Akt-targeted nitroxide-conjugated peptide could be a useful treatment option for controlling vascular proliferation in PAH.
Collapse
|
33
|
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
|
34
|
Tseng V, Sutliff RL, Hart CM. Redox Biology of Peroxisome Proliferator-Activated Receptor-γ in Pulmonary Hypertension. Antioxid Redox Signal 2019; 31:874-897. [PMID: 30582337 PMCID: PMC6751396 DOI: 10.1089/ars.2018.7695] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Peroxisome proliferator-activated receptor-gamma (PPARγ) maintains pulmonary vascular health through coordination of antioxidant defense systems, inflammation, and cellular metabolism. Insufficient PPARγ contributes to pulmonary hypertension (PH) pathogenesis, whereas therapeutic restoration of PPARγ activity attenuates PH in preclinical models. Recent Advances: Numerous studies in the past decade have elucidated the complex mechanisms by which PPARγ in the pulmonary vasculature and right ventricle (RV) protects against PH. The scope of PPARγ-interconnected pathways continues to expand and includes induction of antioxidant genes, transrepression of inflammatory signaling, regulation of mitochondrial biogenesis and bioenergetic integrity, control of cell cycle and proliferation, and regulation of vascular tone through interactions with nitric oxide and endogenous vasoactive molecules. Furthermore, PPARγ interacts with an extensive regulatory network of transcription factors and microRNAs leading to broad impact on cell signaling. Critical Issues: Abundant evidence suggests that targeting PPARγ exerts diverse salutary effects in PH and represents a novel and potentially translatable therapeutic strategy. However, progress has been slowed by an incomplete understanding of how specific PPARγ pathways are critically disrupted across PH disease subtypes and lack of optimal pharmacological ligands. Future Directions: Recent studies indicate that ligand-induced post-translational modifications of the PPARγ receptor differentially induce therapeutic benefits versus adverse side effects of PPARγ receptor activation. Strategies to selectively target PPARγ activity in diseased cells of pulmonary circulation and RV, coupled with development of ligands designed to specifically regulate post-translational PPARγ modifications, may unlock the full therapeutic potential of this versatile master transcriptional and metabolic regulator in PH.
Collapse
Affiliation(s)
- Victor Tseng
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Roy L Sutliff
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - C Michael Hart
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, Georgia.,Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| |
Collapse
|
35
|
Weise-Cross L, Resta TC, Jernigan NL. Redox Regulation of Ion Channels and Receptors in Pulmonary Hypertension. Antioxid Redox Signal 2019; 31:898-915. [PMID: 30569735 PMCID: PMC7061297 DOI: 10.1089/ars.2018.7699] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 12/11/2018] [Indexed: 02/06/2023]
Abstract
Significance: Pulmonary hypertension (PH) is characterized by elevated vascular resistance due to vasoconstriction and remodeling of the normally low-pressure pulmonary vasculature. Redox stress contributes to the pathophysiology of this disease by altering the regulation and activity of membrane receptors, K+ channels, and intracellular Ca2+ homeostasis. Recent Advances: Antioxidant therapies have had limited success in treating PH, leading to a growing appreciation that reductive stress, in addition to oxidative stress, plays a role in metabolic and cell signaling dysfunction in pulmonary vascular cells. Reactive oxygen species generation from mitochondria and NADPH oxidases has substantial effects on K+ conductance and membrane potential, and both receptor-operated and store-operated Ca2+ entry. Critical Issues: Some specific redox changes resulting from oxidation, S-nitrosylation, and S-glutathionylation are known to modulate membrane receptor and ion channel activity in PH. However, many sites of regulation that have been elucidated in nonpulmonary cell types have not been tested in the pulmonary vasculature, and context-specific molecular mechanisms are lacking. Future Directions: Here, we review what is known about redox regulation of membrane receptors and ion channels in PH. Further investigation of the mechanisms involved is needed to better understand the etiology of PH and develop better targeted treatment strategies.
Collapse
Affiliation(s)
- Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C. Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
36
|
Suliman HB, Nozik-Grayck E. Mitochondrial Dysfunction: Metabolic Drivers of Pulmonary Hypertension. Antioxid Redox Signal 2019; 31:843-857. [PMID: 30604624 PMCID: PMC6751393 DOI: 10.1089/ars.2018.7705] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Pulmonary hypertension (PH) is a progressive disease characterized by pulmonary vascular remodeling and lung vasculopathy. The disease displays progressive dyspnea, pulmonary artery uncoupling and right ventricular (RV) dysfunction. The overall survival rate is ranging from 28-72%. Recent Advances: The molecular events that promote the development of PH are complex and incompletely understood. Metabolic impairment has been proposed to contribute to the pathophysiology of PH with evidence for mitochondrial dysfunction involving the electron transport chain proteins, antioxidant enzymes, apoptosis regulators, and mitochondrial quality control. Critical Issues: It is vital to characterize the mechanisms by which mitochondrial dysfunction contribute to PH pathogenesis. This review focuses on the currently available publications that supports mitochondrial mechanisms in PH pathophysiology. Future Directions: Further studies of these metabolic mitochondrial alterations in PH could be viable targets of diagnostic and therapeutic intervention.
Collapse
Affiliation(s)
- Hagir B Suliman
- Department of Anesthesiology, Duke University Medical Centers, Durham, North Carolina
| | - Eva Nozik-Grayck
- Department of Pediatrics, Cardiovascular Pulmonary Research Labs and Pediatric Critical Care Medicine, University of Colorado Denver, Aurora, Colorado
| |
Collapse
|
37
|
Expression of antioxidant genes in broiler chickens fed nettle ( Urtica dioica) and its link with pulmonary hypertension. ACTA ACUST UNITED AC 2019; 5:264-269. [PMID: 31528728 PMCID: PMC6737495 DOI: 10.1016/j.aninu.2019.04.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022]
Abstract
Nettle (Urtica dioica) contains a wide range of chemical constituents that confer a strong antioxidant capacity to the plant. The present study was to investigate the antioxidant gene expression and pulmonary hypertensive responses of broiler chickens to U. dioica. A total of 240 one-d-old broilers (Ross 308) were randomly assigned to 4 dietary levels of U. dioica (0, 0.5%, 1% and 1.5%). Birds were reared for 6 wk in a high altitude region (2,100 m). The results showed a significant relative overexpression (target gene/β-actin as the arbitrary unit) of catalase (CAT) and superoxide dismutase 1 (SOD1) in the liver and lung of the chickens fed U. dioica. Lipid peroxidation was significantly suppressed, as reflected in reduced circulatory concentrations of malondialdehyde (MDA) in the birds fed U. dioica. These birds also had significantly (P < 0.05) higher serum nitric oxide (NO) concentrations than those in the control group. Feeding U. dioica at 1% and 1.5% also attenuated the right ventricular hypertrophy (reflected in the lower right to total ventricular weight ratio), which was associated with a significant lower rate of mortality from pulmonary hypertension syndrome. Feeding U. dioica led to an upregulation of hepatic and pulmonary antioxidant genes.
Collapse
|
38
|
Yang YD, Li MM, Xu G, Zhang EL, Chen J, Sun B, Chen DW, Gao YQ. Targeting mitochondria-associated membranes as a potential therapy against endothelial injury induced by hypoxia. J Cell Biochem 2019; 120:18967-18978. [PMID: 31241212 DOI: 10.1002/jcb.29220] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/04/2019] [Indexed: 02/06/2023]
Abstract
Mitochondrial dysfunction plays a principal role in hypoxia-induced endothelial injury, which is involved in hypoxic pulmonary hypertension and ischemic cardiovascular diseases. Recent studies have identified mitochondria-associated membranes (MAMs) that modulate mitochondrial function under a variety of pathophysiological conditions such as high-fat diet-mediated insulin resistance, hypoxia reoxygenation-induced myocardial death, and hypoxia-evoked vascular smooth muscle cell proliferation. However, the role of MAMs in hypoxia-induced endothelial injury remains unclear. To explore this further, human umbilical vein endothelial cells and human pulmonary artery endothelial cells were exposed to hypoxia (1% O2 ) for 24 hours. An increase in MAM formation was uncovered by immunoblotting and immunofluorescence. Then, we performed small interfering RNA transfection targeted to MAM constitutive proteins and explored the biological effects. Knockdown of MAM constitutive proteins attenuated hypoxia-induced elevation of mitochondrial Ca2+ and repressed mitochondrial impairment, leading to an increase in mitochondrial membrane potential and ATP production and a decline in reactive oxygen species. Then, we found that MAM disruption mitigated cell apoptosis and promoted cell survival. Next, other protective effects, such as those pertaining to the repression of inflammatory response and the promotion of NO synthesis, were investigated. With the disruption of MAMs under hypoxia, inflammatory molecule expression was repressed, and the eNOS-NO pathway was enhanced. This study demonstrates that the disruption of MAMs might be of therapeutic value for treating endothelial injury under hypoxia, suggesting a novel strategy for preventing hypoxic pulmonary hypertension and ischemic injuries.
Collapse
Affiliation(s)
- Yi-Dong Yang
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China.,Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Man-Man Li
- Genetics Laboratory, Hubei Maternal and Child Health Hospital, Wuhan, China
| | - Gang Xu
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China.,Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Er-Long Zhang
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China.,Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Jian Chen
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China.,Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - Binda Sun
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China.,Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| | - De-Wei Chen
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China.,Key Laboratory of High Altitude Medicine, PLA, Chongqing, China.,Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yu-Qi Gao
- Institute of Medicine and Hygienic Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China.,Key Laboratory of High Altitude Medicine, PLA, Chongqing, China
| |
Collapse
|
39
|
Role of Gender in Regulation of Redox Homeostasis in Pulmonary Arterial Hypertension. Antioxidants (Basel) 2019; 8:antiox8050135. [PMID: 31100969 PMCID: PMC6562572 DOI: 10.3390/antiox8050135] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/18/2019] [Accepted: 05/09/2019] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is one of the diseases with a well-established gender dimorphism. The prevalence of PAH is increased in females with a ratio of 4:1, while poor survival prognosis is associated with the male gender. Nevertheless, the specific contribution of gender in disease development and progression is unclear due to the complex nature of the PAH. Oxidative and nitrosative stresses are important contributors in PAH pathogenesis; however, the role of gender in redox homeostasis has been understudied. This review is aimed to overview the possible sex-specific mechanisms responsible for the regulation of the balance between oxidants and antioxidants in relation to PAH pathobiology.
Collapse
|
40
|
Huetsch JC, Suresh K, Shimoda LA. Regulation of Smooth Muscle Cell Proliferation by NADPH Oxidases in Pulmonary Hypertension. Antioxidants (Basel) 2019; 8:antiox8030056. [PMID: 30841544 PMCID: PMC6466559 DOI: 10.3390/antiox8030056] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 02/07/2023] Open
Abstract
Hyperproliferation of pulmonary arterial smooth muscle cells is a key component of vascular remodeling in the setting of pulmonary hypertension (PH). Numerous studies have explored factors governing the changes in smooth muscle cell phenotype that lead to the increased wall thickness, and have identified various potential candidates. A role for reactive oxygen species (ROS) has been well documented in PH. ROS can be generated from a variety of sources, including mitochondria, uncoupled nitric oxide synthase, xanthine oxidase, and reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. In this article, we will review recent data supporting a role for ROS generated from NADPH oxidases in promoting pulmonary arterial smooth muscle cell proliferation during PH.
Collapse
Affiliation(s)
- John C Huetsch
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA.
| | - Karthik Suresh
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA.
| | - Larissa A Shimoda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA.
| |
Collapse
|
41
|
Murtaza G, Paddenberg R, Pfeil U, Goldenberg A, Mermer P, Kummer W. Hypoxia-induced pulmonary vasoconstriction of intra-acinar arteries is impaired in NADPH oxidase 4 gene-deficient mice. Pulm Circ 2018; 8:2045894018808240. [PMID: 30284509 PMCID: PMC6236867 DOI: 10.1177/2045894018808240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We show that genetic deficiency of the reactive oxygen species generating enzyme
NADPH oxidase 4 (NOX4) impairs hypoxic pulmonary vasoconstriction in small
(25–40 µm) intra-acinar, but not pre-acinar, arteries in murine precision cut
lung slices. These data suggest an involvement of NOX4 in ventilation-perfusion
matching at the acinar level.
Collapse
Affiliation(s)
- Ghulam Murtaza
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany.,2 Department of Zoology, University of Gujrat, Gujrat, Pakistan
| | - Renate Paddenberg
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Uwe Pfeil
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Anna Goldenberg
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Petra Mermer
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | - Wolfgang Kummer
- 1 Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany.,3 German Center for Lung Research, Excellence Cluster Cardio-Pulmonary System, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
42
|
Guo X, Fan Y, Cui J, Hao B, Zhu L, Sun X, He J, Yang J, Dong J, Wang Y, Liu X, Chen J. NOX4 expression and distal arteriolar remodeling correlate with pulmonary hypertension in COPD. BMC Pulm Med 2018; 18:111. [PMID: 29986678 PMCID: PMC6038356 DOI: 10.1186/s12890-018-0680-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/25/2018] [Indexed: 12/26/2022] Open
Abstract
Background Pulmonary hypertension (PH) in chronic obstructive pulmonary disease (COPD) is suggested as the consequence of emphysematous destruction of vascular bed and hypoxia of pulmonary microenvironment, mechanisms underpinning its pathogenesis however remain elusive. The dysregulated expression of nicotinamide adenine dinucleotide phosphate (NADPH)-oxidases and superoxide generation by pulmonary vasculatures have significant implications in the hypoxia-induced PH. Methods In this study, the involvement of NADPH oxidase subunit 4 (NOX4) in pulmonary arteriolar remodeling of PH in COPD was investigated by ascertaining the morphological alteration of pulmonary arteries and pulmonary blood flow using cardiac magnetic resonance imaging (cMRI), and the expression and correlation of NOX4 with pulmonary vascular remodeling and pulmonary functions in COPD lungs. Results Results demonstrated that an augmented expression of NOX4 was correlated with the increased volume of pulmonary vascular wall in COPD lung. While the volume of distal pulmonary arteries was inversely correlated with pulmonary functions, despite it was positively associated with the main pulmonary artery distensibility, right ventricular myocardial mass end-systolic and right ventricular myocardial mass end-diastolic in COPD. In addition, an increased malondialdehyde and a decreased superoxide dismutase were observed in sera of COPD patients. Mechanistically, the abundance of NOX4 and production of reactive oxygen species (ROS) in pulmonary artery smooth muscle cells could be dynamically induced by transforming growth factor-beta (TGF-β), which in turn led pulmonary arteriolar remodeling in COPD lungs. Conclusion These results suggest that the NOX4-derived ROS production may play a key role in the development of PH in COPD by promoting distal pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Xiaotong Guo
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, Ningxia, 750004, People's Republic of China
| | - Yuchun Fan
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, Ningxia, 750004, People's Republic of China
| | - Jieda Cui
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, Ningxia, 750004, People's Republic of China.,Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Binwei Hao
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, Ningxia, 750004, People's Republic of China
| | - Li Zhu
- Department of Radiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xiao Sun
- Department of Radiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jinxi He
- Department of Thoracic Surgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jiali Yang
- Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Jianda Dong
- Department of Pathology, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yanyang Wang
- Department of Radiotherapy, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Xiaoming Liu
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, Ningxia, 750004, People's Republic of China. .,Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| | - Juan Chen
- Department of Pulmonary and Critical Care Medicine, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan, Ningxia, 750004, People's Republic of China.
| |
Collapse
|
43
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
44
|
Recurrent inhibition of mitochondrial complex III induces chronic pulmonary vasoconstriction and glycolytic switch in the rat lung. Respir Res 2018; 19:69. [PMID: 29685148 PMCID: PMC5914012 DOI: 10.1186/s12931-018-0776-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/12/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a fatal disease; however, the mechanisms directly involved in triggering and the progression of PAH are not clear. Based on previous studies that demonstrated a possible role of mitochondrial dysfunction in the pathogenesis of PAH, we investigated the effects of chronic inhibition of mitochondrial function in vivo in healthy rodents. METHODS Right ventricle systolic pressure (RVSP) was measured in female rats at baseline and up to 24 days after inhibition of mitochondrial respiratory Complex III, induced by Antimycin A (AA, 0.35 mg/kg, given three times starting at baseline and then days 3 and 6 as a bolus injection into the right atrial chamber). RESULTS Rodents exposed to AA demonstrated sustained increases in RVSP from days 6 through 24. AA-exposed rodents also possessed a progressive increase in RV end-diastolic pressure but not RV hypertrophy, which may be attributed to either early stages of PAH development or to reduced RV contractility due to inhibition of myocardial respiration. Protein nitration levels in plasma were positively correlated with PAH development in AA-treated rats. This finding was strongly supported by results obtained from PAH humans where plasma protein nitration levels were correlated with markers of PAH severity in female but not male PAH patients. Based on previously reported associations between increased nitric oxide production levels with female gender, we speculate that in females with PAH mitochondrial dysfunction may represent a more deleterious form, in part, due to an increased nitrosative stress development. Indeed, the histological analysis of AA treated rats revealed a strong perivascular edema, a marker of pulmonary endothelial damage. Finally, AA treatment was accompanied by a severe metabolic shift toward glycolysis, a hallmark of PAH pathology. CONCLUSIONS Chronic mitochondrial dysfunction induces the combination of vascular damage and metabolic reprogramming that may be responsible for PAH development. This mechanism may be especially important in females, perhaps due to an increased NO production and nitrosative stress development.
Collapse
|
45
|
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
|
46
|
From Physiological Redox Signalling to Oxidant Stress. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:335-342. [PMID: 29047097 DOI: 10.1007/978-3-319-63245-2_21] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oxidant stress is strongly associated with cardiovascular disease, including pulmonary hypertension, but antioxidant therapies have so far proven ineffective. This is partly due to a lack of understanding of the key role played by reactive oxygen species (ROS) in physiological cell signalling, and partly to the complex interrelationships between generators of ROS (e.g. mitochondria and NADPH oxidases, NOX), cellular antioxidant systems and indeed Ca2+ signalling. At physiological levels ROS reversibly affect the function of numerous enzymes and transcription factors, most often via oxidation of specific protein thiols. Importantly, they also affect pathways that promote ROS generation by NOX or mitochondria (ROS-induced ROS release), which has an inherent propensity for positive feedback and uncontrolled oxidant production. The reason this does not occur under normal conditions reflects in part a high level of compartmentalisation of ROS signalling within the cell, akin to that for Ca2+. This article considers the physiological processes which regulate NOX and mitochondrial ROS production and degradation and their interactions with each other and Ca2+ signalling pathways, and discusses how loss of spatiotemporal constraints and activation of positive feedback pathways may impact on their dysregulation in pulmonary hypertension.
Collapse
|
47
|
Pak O, Scheibe S, Esfandiary A, Gierhardt M, Sydykov A, Logan A, Fysikopoulos A, Veit F, Hecker M, Kroschel F, Quanz K, Erb A, Schäfer K, Fassbinder M, Alebrahimdehkordi N, Ghofrani HA, Schermuly RT, Brandes RP, Seeger W, Murphy MP, Weissmann N, Sommer N. Impact of the mitochondria-targeted antioxidant MitoQ on hypoxia-induced pulmonary hypertension. Eur Respir J 2018; 51:1701024. [PMID: 29419444 DOI: 10.1183/13993003.01024-2017] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 01/10/2018] [Indexed: 12/18/2022]
Abstract
Increased mitochondrial reactive oxygen species (ROS), particularly superoxide, have been suggested to mediate hypoxic pulmonary vasoconstriction (HPV), chronic hypoxia-induced pulmonary hypertension and right ventricular remodelling.We determined ROS in acute and chronic hypoxia, and investigated the effect of the mitochondria-targeted antioxidant MitoQ under these conditions.The effect of MitoQ or its inactive carrier substance, decyltriphenylphosphonium, on acute HPV (1% O2 for 10 min) was investigated in isolated blood-free perfused mouse lungs. Mice exposed to chronic hypoxia (10% O2 for 4 weeks) or after banding of the main pulmonary artery were treated with MitoQ or decyltriphenylphosphonium (50 mg·kg-1·day-1).Total cellular superoxide and mitochondrial ROS levels were increased in pulmonary artery smooth muscle cells but decreased in pulmonary fibroblasts in acute hypoxia. MitoQ significantly inhibited HPV and acute hypoxia-induced rise in superoxide concentration. ROS was decreased in pulmonary artery smooth muscle cells, while it increased in the right ventricle after chronic hypoxia. Correspondingly, MitoQ did not affect the development of chronic hypoxia-induced pulmonary hypertension but attenuated right ventricular remodelling after chronic hypoxia as well as after pulmonary arterial banding.Increased mitochondrial ROS of pulmonary artery smooth muscle cells mediate acute HPV, but not chronic hypoxia-induced pulmonary hypertension. MitoQ may be beneficial under conditions of exaggerated acute HPV.
Collapse
Affiliation(s)
- Oleg Pak
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- These two authors contributed equally to this work
| | - Susan Scheibe
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- These two authors contributed equally to this work
| | - Azadeh Esfandiary
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Mareike Gierhardt
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Akylbek Sydykov
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | | | - Athanasios Fysikopoulos
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Florian Veit
- Legal Medicine, University of Giessen, Giessen, Germany
| | - Matthias Hecker
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Florian Kroschel
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Karin Quanz
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Alexandra Erb
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Katharina Schäfer
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Mirja Fassbinder
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Nasim Alebrahimdehkordi
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Hossein A Ghofrani
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, German Center for Cardiovascular Research (DZHK), Partner Site Rhein-Main, Frankfurt am Main, Germany
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- These two authors contributed equally to this work
| | - Natascha Sommer
- Excellence Cluster Cardio-Pulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, Giessen, Germany
- These two authors contributed equally to this work
| |
Collapse
|
48
|
Bonnet S, Boucherat O. The ROS controversy in hypoxic pulmonary hypertension revisited. Eur Respir J 2018; 51:1800276. [PMID: 29519907 DOI: 10.1183/13993003.00276-2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/13/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Sébastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
- Dept of Medicine, Université Laval, Québec, QC, Canada
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
- Dept of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
49
|
He X, Song S, Ayon RJ, Balisterieri A, Black SM, Makino A, Wier WG, Zang WJ, Yuan JXJ. Hypoxia selectively upregulates cation channels and increases cytosolic [Ca 2+] in pulmonary, but not coronary, arterial smooth muscle cells. Am J Physiol Cell Physiol 2018; 314:C504-C517. [PMID: 29351410 DOI: 10.1152/ajpcell.00272.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ca2+ signaling, particularly the mechanism via store-operated Ca2+ entry (SOCE) and receptor-operated Ca2+ entry (ROCE), plays a critical role in the development of acute hypoxia-induced pulmonary vasoconstriction and chronic hypoxia-induced pulmonary hypertension. This study aimed to test the hypothesis that chronic hypoxia differentially regulates the expression of proteins that mediate SOCE and ROCE [stromal interacting molecule (STIM), Orai, and canonical transient receptor potential channel TRPC6] in pulmonary (PASMC) and coronary (CASMC) artery smooth muscle cells. The resting cytosolic [Ca2+] ([Ca2+]cyt) and the stored [Ca2+] in the sarcoplasmic reticulum were not different in CASMC and PASMC. Seahorse measurement showed a similar level of mitochondrial bioenergetics (basal respiration and ATP production) between CASMC and PASMC. Glycolysis was significantly higher in PASMC than in CASMC. The amplitudes of cyclopiazonic acid-induced SOCE and OAG-induced ROCE in CASMC are slightly, but significantly, greater than in PASMC. The frequency and the area under the curve of Ca2+ oscillations induced by ATP and histamine were also larger in CASMC than in PASMC. Na+/Ca2+ exchanger-mediated increases in [Ca2+]cyt did not differ significantly between CASMC and PASMC. The basal protein expression levels of STIM1/2, Orai1/2, and TRPC6 were higher in CASMC than in PASMC, but hypoxia (3% O2 for 72 h) significantly upregulated protein expression levels of STIM1/STIM2, Orai1/Orai2, and TRPC6 and increased the resting [Ca2+]cyt only in PASMC, but not in CASMC. The different response of essential components of store-operated and receptor-operated Ca2+ channels to hypoxia is a unique intrinsic property of PASMC, which is likely one of the important explanations why hypoxia causes pulmonary vasoconstriction and induces pulmonary vascular remodeling, but causes coronary vasodilation.
Collapse
Affiliation(s)
- Xi He
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi Province, China.,Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Shanshan Song
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Ramon J Ayon
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Angela Balisterieri
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Stephen M Black
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - Ayako Makino
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - W Gil Wier
- Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - Wei-Jin Zang
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shannxi Province, China
| | - Jason X-J Yuan
- Division of Translational and Regenerative Medicine, Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| |
Collapse
|
50
|
Mistriotis P, Andreadis ST. Vascular aging: Molecular mechanisms and potential treatments for vascular rejuvenation. Ageing Res Rev 2017; 37:94-116. [PMID: 28579130 DOI: 10.1016/j.arr.2017.05.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
Aging is the main risk factor contributing to vascular dysfunction and the progression of vascular diseases. In this review, we discuss the causes and mechanisms of vascular aging at the tissue and cellular level. We focus on Endothelial Cell (EC) and Smooth Muscle Cell (SMC) aging due to their critical role in mediating the defective vascular phenotype. We elaborate on two categories that contribute to cellular dysfunction: cell extrinsic and intrinsic factors. Extrinsic factors reflect systemic or environmental changes which alter EC and SMC homeostasis compromising vascular function. Intrinsic factors induce EC and SMC transformation resulting in cellular senescence. Replenishing or rejuvenating the aged/dysfunctional vascular cells is critical to the effective repair of the vasculature. As such, this review also elaborates on recent findings which indicate that stem cell and gene therapies may restore the impaired vascular cell function, reverse vascular aging, and prolong lifespan.
Collapse
Affiliation(s)
- Panagiotis Mistriotis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA
| | - Stelios T Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260-4200, USA; Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA.
| |
Collapse
|