1
|
Meng L, Liu S, Luo J, Tu Y, Li T, Li P, Yu J, Shi L. Oxidative stress and reactive oxygen species in otorhinolaryngological diseases: insights from pathophysiology to targeted antioxidant therapies. Redox Rep 2025; 30:2458942. [PMID: 39894944 PMCID: PMC11792148 DOI: 10.1080/13510002.2025.2458942] [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] [Indexed: 02/04/2025] Open
Abstract
Oxidative stress, characterized by an imbalance between excessive reactive oxygen species (ROS) production and impaired antioxidant defenses, is closely linked to the pathogenesis of various otorhinolaryngological disorders. Mitochondria, as the primary site of cellular energy production, play a crucial role in modulating oxidative stress. Mitochondrial dysfunction exacerbates ROS generation, leading to cellular damage and inflammatory responses. In otorhinolaryngological diseases, oxidative stress is strongly associated with conditions such as hearing loss, allergic rhinitis, and chronic sinusitis, where oxidative damage and tissue inflammation are key pathological features. Recent studies have highlighted the potential of antioxidant therapies to mitigate oxidative stress and restore homeostasis, offering promising avenues for alleviating symptoms in these diseases. However, despite the encouraging results from early-stage research, the clinical efficacy of antioxidant interventions remains to be fully established. This review provides an overview of the role of oxidative stress in otorhinolaryngological diseases and evaluates the therapeutic potential of antioxidant strategies.
Collapse
Affiliation(s)
- Linghui Meng
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, People’s Republic of China
| | - Shengyang Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, People’s Republic of China
| | - Jinfeng Luo
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, People’s Republic of China
| | - Yanyi Tu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, People’s Republic of China
| | - Tao Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, People’s Republic of China
| | - Ping Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, People’s Republic of China
| | - Jinzhuang Yu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, People’s Republic of China
| | - Li Shi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, People’s Republic of China
- Department of Allergy and Immunology, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, People's Republic of China
- The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
2
|
Gupta A, Zaghloul N, Thulasingam SK, Robbins IR, Gupta G, Bader J, Garcia JG, Ahmed M. Tailored CD4+ lymphocytes expressing human CHAT protein as a novel vasodilator in attenuating RV pressure in PAH animal model. Transl Res 2025; 278:22-35. [PMID: 39961494 DOI: 10.1016/j.trsl.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/01/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
ChAT-expressing T cells represent ∼0.01 % of total circulating T lymphocytes in adult wild-type mice. However, we previously reported that systemic infusion of ChAT+ve Jurkat T cells into adult mice elicits vasodilation and instantaneous decline in the mean systolic blood pressure, suggesting potential as a biologic therapeutic to attenuate pathologic increases in pulmonary arterial pressures. We now report that ChAT gene-expressing Jurkat cells dose-dependently decrease right ventricular systolic pressures (RVSP) in hypoxic mice and that transgenic mice with ChAT KO restricted to endothelial cells (KO END/ChAT-/-) exhibit significantly elevated pulmonary arterial pressure and peripheral systemic resistance (compared to WT mice). To rigorously characterize the role of CD4 ChAT+ T cells in regulating pulmonary arterial hypertension (PAH) hemodynamics and molecular signatures, we infused CD4+ ChAT+ve cells (0.5 to 2.0 million cells/animal) into adult PAH mice and noted significant reductions in RVSP within 2-3 min post injection (∼ 50 % reduction). The tailored tail vein injection effect was sustained until the animal was euthanized (30-40 min). Mice KO END/ChAT-/-showed a significant and severe hypoxia-induced PAH phenotype compared to WT adult mice. Tail vein injection of biologically active CD4 ChAT+ve cells into either KO END/ChAT-/-mice with hypoxia-induced PAH or into adult rats with hypoxia/Sugen-induced PAH resulted in significant attenuation of RVP elevations. RNA seq data analysis of human pulmonary endothelial cells (HPAECs) incubated with CD4 ChAT+ve T cells showed significant differential regulation of pathways involved in systemic and pulmonary pressure regulation, NO synthesis/regulation, antioxidant expression, and vasodilation. In conclusion, CD4 ChAT+ve T cells have a unique, vasodilating innate immunity mechanism to augment nitric oxide release and potentially mitigate molecular and genetic pathways involved in PAH pathogenesis.
Collapse
Affiliation(s)
- Akash Gupta
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Nahla Zaghloul
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States; Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | | | | | - Geetanjali Gupta
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Jad Bader
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States
| | - Joe Gn Garcia
- Center for Inflammation Science and Systems Medicine, University of Florida Wertheim Scripps Research Institute, Jupiter, FL, United States
| | - Mohamed Ahmed
- Department of Pediatrics, University of Arizona, Tucson, AZ, United States; Department of Pediatrics, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
3
|
Jiang P, Huang H, Liu Z, Xiang G, Wu X, Hao S, Li S. STAT6 deficiency mitigates the severity of pulmonary arterial hypertension caused by chronic intermittent hypoxia by suppressing Th2-inducing cytokines. Respir Res 2025; 26:13. [PMID: 39806384 PMCID: PMC11731530 DOI: 10.1186/s12931-024-03062-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is frequently associated with increased incidence and mortality of pulmonary hypertension (PH). The immune response contributes to pulmonary artery remodeling and OSA-related diseases. The immunologic factors linked to OSA-induced PH are not well understood. STAT6 is crucial in the signaling pathway that modulates immune response. However, the status of phosphorylated STAT6 (p-STAT6) in an OSA-induced PH mouse model remains largely unexplored. METHODS Chronic intermittent hypoxia (CIH) plays a crucial role in the progression of OSA. This study utilized a in vivo CIH model to examine the role of STAT6 in CIH-induced PH. RESULTS CIH mice exhibited pulmonary artery remodeling and pulmonary hypertension, indicated by increased right ventricular systolic pressure (RVSP), higher right ventricular to left ventricular plus septum (RV/LV + S) ratios, and significant morphological alterations compared to normoxic (Nor) mice. Increased p-STAT6 in the lungs and elevated p-STAT6 + IL-4 + producing T cells in CIH mice. STAT6 deficiency (STAT6-/-) improved PH and PA remodeling in CIH-induced PH mouse models.STAT6 deficiency impaired the T helper 2 (Th2) immune response, affecting IL-4 and IL-13 secretion. IL-4, rather than IL-13, activated STAT6 in human pulmonary artery smooth muscle cells (hPASMCs). STAT6 knockdown decreased the proliferation in IL-4 treated hPASMCs. CONCLUSION These findings exhibit the critical role of STAT6 in the pathogenesis of CIH induced PH by regulating Th2 immune response.STAT6 could be a significant therapeutic target for OSA-related PH.
Collapse
Affiliation(s)
- Pan Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- The Nutrition Department at Zhongshan Hospital, Fudan University, Shanghai, China
- The Nutrition Department, QingPu District Central Hospital, Shanghai, 200032, China
| | - Huai Huang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zilong Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guiling Xiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- The Critical Care Medicine Department at Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaodan Wu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Clinical Center for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Shengyu Hao
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- The Critical Care Medicine Department at Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Shanqun Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Clinical Center for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
4
|
Zhang X, Wang Y, Pan Z, Hu K. New insights from integrated bioinformatics analysis: the role of circadian rhythm disruption and immune infiltration in obstructive sleep apnea disease. Front Immunol 2023; 14:1273114. [PMID: 38169659 PMCID: PMC10758485 DOI: 10.3389/fimmu.2023.1273114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Background Circadian rhythm disruption and immune infiltration are both closely associated with the development of Obstructive sleep apnea (OSA) disease and a variety of cardiovascular and neurological complications, but their interactions with OSA disease are not clear. In this study, we used bioinformatics to investigate the roles of circadian rhythm disruption and immune microenvironments in OSA. Methods We analyzed differential genes and their associated functional pathways in the circadian rhythm-associated OSA dataset, then regrouped OSA samples using the differential genes and explored differences in immune cell infiltration between the two different subgroups. Meanwhile, we used two machine learning algorithms to further define circadian rhythm-related signature genes and to explore the relationship between key genes and immune cell infiltration. Finally, we searched for the transcription factors of the key differential gene JUN. Results We screened 15 circadian rhythm-related differential genes in the OSA-related dataset and further defined 3 signature genes by machine learning algorithms. Immunoassays showed a significant increase in resting mast cell infiltration and a decrease in monocyte infiltration in the OSA group. The results of our animal experiments also confirmed that the expression of these 3 key genes, as well as the immune cell infiltration, showed a trend consistent with the results of the bioinformatics analysis. Conclusions In conclusion, this study reveals the interaction between circadian rhythm disruption and immune infiltration in OSA, providing new insights into the potential pathogenesis of OSA.
Collapse
|
5
|
Pan Z, Wu X, Zhang X, Hu K. Phosphodiesterase 4B activation exacerbates pulmonary hypertension induced by intermittent hypoxia by regulating mitochondrial injury and cAMP/PKA/p-CREB/PGC-1α signaling. Biomed Pharmacother 2023; 158:114095. [PMID: 36495666 DOI: 10.1016/j.biopha.2022.114095] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Proliferation of smooth muscle cells, oxidative stress, and pulmonary vasoconstriction resulting from intermittent hypoxia (IH) facilitate pulmonary hypertension (PH) in patients with obstructive sleep apnea. The role of Phosphodiesterase 4 B (PDE4B) in PH has not yet been established. Herein, we investigated whether PDE4B inhibition ameliorates experimental PH by modulating cAMP signaling. We performed an integrative analysis of PDE4B expression in Gene Expression Omnibus datasets, experimental IH-induced rat PH samples, and IH-induced pulmonary arterial smooth muscle cells (PASMCs). PDE4B expression was modulated using siRNA in vitro and a specific adeno-associated virus serotype 1 in vivo. In the databases of mouse models of IH-induced and sustained hypoxia-induced PH and in a rat model of six weeks of IH, the expression of PDE4B was up-regulated. Inhibition of PDE4B attenuated IH-induced pulmonary vascular remodeling and right ventricular hypertrophy. Our results also showed that PDE4B deficiency inhibited IH-induced proliferation of PASMCs with less mitochondrial reactive oxygen species and mitochondrial damage. Meanwhile, IH induced an increase in ATF4, which positively regulated the expression of PDE4B through transcription, and inhibition of ATF4 exerted effects similar to those of PDE4B inhibition. Mechanistically, downregulating the expression of PDE4B resulted in the activation of the cAMP/PKA/p-CREB/PGC-1α pathway in PASMCs after IH. Taken together, our present study provides evidence that inhibition of PDE4B attenuates IH-induced PH by regulating cAMP signaling.
Collapse
Affiliation(s)
- Zhou Pan
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaofeng Wu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xinyue Zhang
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ke Hu
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
6
|
Hu S, Wang L, Xu Y, Li F, Wang T. Disulfiram attenuates hypoxia-induced pulmonary hypertension by inhibiting GSDMD cleavage and pyroptosis in HPASMCs. Respir Res 2022; 23:353. [PMID: 36527086 PMCID: PMC9756478 DOI: 10.1186/s12931-022-02279-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is characterized by progressive pulmonary arterial remodelling, associated with different severities of inflammation and altered immune processes. Disulfiram eliminates the formation of N-gasdermin D (GSDMD) plasma membrane pores to prevent pyroptosis. Pyroptosis is a form of lytic cell death characterized by inflammasome activation and proinflammatory cytokine release that acts in the development of PH. We sought to investigate whether disulfiram could alleviate hypoxia-induced PH by inhibiting pyroptosis. METHODS To investigate whether disulfiram alleviates the progression of pulmonary hypertension, rodents were exposed to chronic hypoxia (10% oxygen, 4 weeks) to induce PH. The severity of PH was assessed by measuring right ventricular systolic pressure, mean pulmonary artery pressure, and the degree of right ventricular hypertrophy. Western blotting was used to measure proteins associated with the pyroptosis pathway, and ELISA was performed to measure the secretion of IL-18 and IL-1β, both of which are the primary methods for assessing pyroptosis. RESULTS IL-18 and IL-1β concentrations were higher in patients with PH than in normal controls. Disulfiram suppressed the progression of PH in mice and rats through the alleviation of pulmonary arterial remodelling. Pyroptosis-related proteins and the inflammasome were activated in rodent models of PH. Disulfiram inhibited the processing of GSDMD into N-GSDMD and attenuated the secretion of IL-1β and IL18. In vivo experiments showed that disulfiram also inhibited lytic death in HPASMCs. CONCLUSIONS Disulfiram treatment reduces PH progression through suppressing vascular remodelling by inhibiting GSDMD cleavage and pyroptosis. It might become a novel therapeutic option for the treatment of PH.
Collapse
Affiliation(s)
- Shunlian Hu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lu Wang
- Department of Respiratory and Critical Care Medicine, Miyun Teaching Hospital of Capital Medical University, Beijing, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Miyun District, Beijing, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Beijing Miyun Hospital, Beijing, People's Republic of China
| | - Yahan Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fajiu Li
- The Sixth Hospital of Wuhan City, Affiliated Hospital of Jianghan University, Beijing, People's Republic of China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- The Center for Biomedical Research, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
7
|
Scott TE, Qin CX, Drummond GR, Hobbs AJ, Kemp-Harper BK. Innovative Anti-Inflammatory and Pro-resolving Strategies for Pulmonary Hypertension: High Blood Pressure Research Council of Australia Award 2019. Hypertension 2021; 78:1168-1184. [PMID: 34565184 DOI: 10.1161/hypertensionaha.120.14525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pulmonary hypertension is a rare, ostensibly incurable, and etiologically diverse disease with an unacceptably high 5-year mortality rate (≈50%), worse than many cancers. Irrespective of pathogenic origin, dysregulated immune processes underlie pulmonary hypertension pathobiology, particularly pertaining to pulmonary vascular remodeling. As such, a variety of proinflammatory pathways have been mooted as novel therapeutic targets. One such pathway involves the family of innate immune regulators known as inflammasomes. In addition, a new and emerging concept is differentiating between anti-inflammatory approaches versus those that promote pro-resolving pathways. This review will briefly introduce inflammasomes and examine recent literature concerning their role in pulmonary hypertension. Moreover, it will explore the difference between inflammation-suppressing and pro-resolution approaches and how this links to inflammasomes. Finally, we will investigate new avenues for targeting inflammation in pulmonary hypertension via more targeted anti-inflammatory or inflammation resolving strategies.
Collapse
Affiliation(s)
- Tara E Scott
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute (T.E.S., B.K.K.-H.), Monash University, Parkville, VIC, Australia
- Monash University, Clayton, VIC, Australia and Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences (T.E.S., C.X.Q.), Monash University, Parkville, VIC, Australia
| | - Cheng Xue Qin
- Monash University, Clayton, VIC, Australia and Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences (T.E.S., C.X.Q.), Monash University, Parkville, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (C.X.Q.)
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia (G.R.D.)
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (A.J.H.)
| | - Barbara K Kemp-Harper
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute (T.E.S., B.K.K.-H.), Monash University, Parkville, VIC, Australia
| |
Collapse
|
8
|
Wang ZB, Jiang SL, Liu SB, Peng JB, Hu S, Wang X, Zhuo W, Liu T, Guo JW, Zhou HH, Yang ZQ, Mao XY, Liu ZQ. Metabolomics of Artichoke Bud Extract in Spontaneously Hypertensive Rats. ACS OMEGA 2021; 6:18610-18622. [PMID: 34337201 PMCID: PMC8319930 DOI: 10.1021/acsomega.1c01135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/05/2021] [Indexed: 05/10/2023]
Abstract
Hypertension adversely affects the quality of life in humans across modern society. Studies have attributed increased reactive oxygen species production to the pathophysiology of hypertension. So far, a specific drug to control the disease perfectly has not been developed. However, artichoke, an edible vegetable, plays an essential role in treating many diseases due to its potent antioxidant activities. The objective of this study is to evaluate the effect of artichoke bud extract (ABE) on heart tissue metabolomics of hypertensive rats. Spontaneously hypertensive rats and Wistar-Kyoto (WKY) rats were divided into six groups, then exposed to different doses comprising ABE, Enalapril Maleate, or 1% carboxylmethyl cellulose for 4 weeks. Their blood pressures were recorded at 0, 2, 3, and 4 weeks after the start of the test period. Thereafter, all rats were anesthetized, and blood was collected from their cardiac apexes. Then, we measured the levels for 15 kinds of serum biochemical parameters. An established orthogonal partial least square-discriminant analysis model completed the metabolomic analysis. Hypertensive rats in the ABE group exhibited well-controlled blood pressure, relative to those in the model group. Specifically, artichoke significantly lowered serum levels for total protein (TP), albumin (ALB), and uric acid (UA) in the hypertensive rats. This effect involved the action of eight metabolites, including guanine, 1-methylnicotinamide, p-aminobenzoic acid, NAD, NADH, uridine 5'-monophosphate, adenosine monophosphate, and methylmalonic acid. Collectively, these findings suggest that ABE may play a role in affecting oxidative stress and purine, nicotinate, and nicotinamide metabolism.
Collapse
Affiliation(s)
- Zhi-Bin Wang
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| | - Shi-Long Jiang
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| | - Shao-Bo Liu
- Department
of Pharmacy, Xiangya Hospital, Central South
University, Changsha 410008, P. R. China
| | - Jing-Bo Peng
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| | - Shuo Hu
- Department
of Nuclear Medicine and Key Laboratory of Biological Nanotechnology
of National Health Commission, Xiangya Hospital, Central South University, Changsha 410008, P. R. China
| | - Xu Wang
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| | - Wei Zhuo
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| | - Tong Liu
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| | - Ji-Wei Guo
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| | - Hong-Hao Zhou
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| | - Zhi-Quan Yang
- Department
of Neurosurgery, Xiangya Hospital, Central
South University, Changsha 410008, P. R. China
- . Phone: +86 731 89753845. Fax: +86 731 82354476
| | - Xiao-Yuan Mao
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| | - Zhao-Qian Liu
- Department
of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics,
and National Clinical Research Center for Geriatric Disorders, Xiangya
Hospital, Central South University, Changsha 410008, P. R. China
- Institute
of Clinical Pharmacology, Engineering Research Center for Applied
Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, P. R. China
| |
Collapse
|
9
|
James J, Zemskova M, Eccles CA, Varghese MV, Niihori M, Barker NK, Luo M, Mandarino LJ, Langlais PR, Rafikova O, Rafikov R. Single Mutation in the NFU1 Gene Metabolically Reprograms Pulmonary Artery Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2021; 41:734-754. [PMID: 33297749 PMCID: PMC7837686 DOI: 10.1161/atvbaha.120.314655] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE NFU1 is a mitochondrial iron-sulfur scaffold protein, involved in iron-sulfur assembly and transfer to complex II and LAS (lipoic acid synthase). Patients with the point mutation NFU1G208C and CRISPR/CAS9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9)-generated rats develop mitochondrial dysfunction leading to pulmonary arterial hypertension. However, the mechanistic understanding of pulmonary vascular proliferation due to a single mutation in NFU1 remains unresolved. Approach and Results: Quantitative proteomics of isolated mitochondria showed the entire phenotypic transformation of NFU1G206C rats with a disturbed mitochondrial proteomic landscape, involving significant changes in the expression of 208 mitochondrial proteins. The NFU1 mutation deranged the expression pattern of electron transport proteins, resulting in a significant decrease in mitochondrial respiration. Reduced reliance on mitochondrial respiration amplified glycolysis in pulmonary artery smooth muscle cell (PASMC) and activated GPD (glycerol-3-phosphate dehydrogenase), linking glycolysis to oxidative phosphorylation and lipid metabolism. Decreased PDH (pyruvate dehydrogenase) activity due to the lipoic acid shortage is compensated by increased fatty acid metabolism and oxidation. PASMC became dependent on extracellular fatty acid sources due to upregulated transporters such as CD36 (cluster of differentiation 36) and CPT (carnitine palmitoyltransferase)-1. Finally, the NFU1 mutation produced a dysregulated antioxidant system in the mitochondria, leading to increased reactive oxygen species levels. PASMC from NFU1 rats showed apoptosis resistance, increased anaplerosis, and attained a highly proliferative phenotype. Attenuation of mitochondrial reactive oxygen species by mitochondrial-targeted antioxidant significantly decreased PASMC proliferation. CONCLUSIONS The alteration in iron-sulfur metabolism completely transforms the proteomic landscape of the mitochondria, leading toward metabolic plasticity and redistribution of energy sources to the acquisition of a proliferative phenotype by the PASMC.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Proliferation
- Cells, Cultured
- Cellular Reprogramming
- Energy Metabolism
- Fatty Acids/metabolism
- Female
- Mitochondria, Liver/genetics
- Mitochondria, Liver/metabolism
- Mitochondria, Liver/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Point Mutation
- Proteome
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Rats
Collapse
Affiliation(s)
- Joel James
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Marina Zemskova
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Cody A. Eccles
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Mathews V. Varghese
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Maki Niihori
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Natalie K. Barker
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Moulun Luo
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Lawrence J. Mandarino
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Paul R. Langlais
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Olga Rafikova
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Ruslan Rafikov
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| |
Collapse
|
10
|
Pellegrini C, Martelli A, Antonioli L, Fornai M, Blandizzi C, Calderone V. NLRP3 inflammasome in cardiovascular diseases: Pathophysiological and pharmacological implications. Med Res Rev 2021; 41:1890-1926. [PMID: 33460162 DOI: 10.1002/med.21781] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/30/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
Growing evidence points out the importance of nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome in the pathogenesis of cardiovascular diseases (CVDs), including hypertension, myocardial infarct (MI), ischemia, cardiomyopathies (CMs), heart failure (HF), and atherosclerosis. In this regard, intensive research efforts both in humans and in animal models of CVDs are being focused on the characterization of the pathophysiological role of NLRP3 inflammasome signaling in CVDs. In addition, clinical and preclinical evidence is coming to light that the pharmacological blockade of NLRP3 pathways with drugs, including novel chemical entities as well as drugs currently employed in the clinical practice, biologics and phytochemicals, could represent a suitable therapeutic approach for prevention and management of CVDs. On these bases, the present review article provides a comprehensive overview of clinical and preclinical studies about the role of NLRP3 inflammasome in the pathophysiology of CVDs, including hypertension, MI, ischemic injury, CMs, HF and atherosclerosis. In addition, particular attention has been focused on current evidence on the effects of drugs, biologics, and phytochemicals, targeting different steps of inflammasome signaling, in CVDs.
Collapse
Affiliation(s)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, Pisa, Italy
| | | |
Collapse
|
11
|
Umeda A, Miyagawa K, Mochida A, Takeda H, Takeda K, Okada Y, Gozal D. Effects of Normoxic Recovery on Intima-Media Thickness of Aorta and Pulmonary Artery Following Intermittent Hypoxia in Mice. Front Physiol 2020; 11:583735. [PMID: 33192596 PMCID: PMC7645053 DOI: 10.3389/fphys.2020.583735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/05/2020] [Indexed: 12/31/2022] Open
Abstract
Obstructive sleep apnea (OSA) patients are at risk for increased blood pressure and carotid intima-media thickness (IMT), with pulmonary hypertension and right-sided heart failure potentially developing as well. Chronic intermittent hypoxia (IH) has been used as an OSA model in animals, but its effects on vascular beds have not been evaluated using objective unbiased tools. Previously published and current experimental data in mice exposed to IH were evaluated for IMT in aorta and pulmonary artery (PA) after IH with or without normoxic recovery using software for meta-analysis, Review Manager 5. Because IMT data reports on PA were extremely scarce, atherosclerotic area percentage from lumen data was also evaluated. IH significantly increased IMT parameters in both aorta and PA as illustrated by Forest plots (P < 0.01), which also confirmed that IMT values after normoxic recovery were within the normal range in both vascular beds. One-sided scarce lower areas in Funnel Plots were seen for both aorta and PA indicating the likelihood of significant publication bias. Forest and Funnel plots, which provide unbiased assessments of published and current data, suggest that IH exposures may induce IMT thickening that may be reversed by normoxic recovery in both aorta and PA. In light of the potential likelihood of publication bias, future studies are needed to confirm or refute the findings. In conclusion, OSA may induce IMT thickening (e.g., aorta and/or PA), but the treatment (e.g., nasal continuous positive airway pressure) will likely lead to improvements in such findings.
Collapse
Affiliation(s)
- Akira Umeda
- Department of Respiratory Medicine, International University of Health and Welfare Shioya Hospital, Yaita, Japan
| | - Kazuya Miyagawa
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, Otawara, Japan
| | - Atsumi Mochida
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, Otawara, Japan
| | - Hiroshi Takeda
- Department of Pharmacology, School of Pharmacy, International University of Health and Welfare, Otawara, Japan
| | - Kotaro Takeda
- Faculty of Rehabilitation, School of Healthcare, Fujita Health University, Toyoake, Japan
| | - Yasumasa Okada
- Department of Internal Medicine, National Hospital Organization Murayama Medical Center, Musashimurayama, Japan
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, MU Women's and Children's Hospital, University of Missouri, Columbia, MO, United States
| |
Collapse
|
12
|
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: 62] [Impact Index Per Article: 12.4] [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
|