1
|
Mu B, Zeng Y, Luo L, Wang K. Oxidative stress-mediated protein sulfenylation in human diseases: Past, present, and future. Redox Biol 2024; 76:103332. [PMID: 39217848 PMCID: PMC11402764 DOI: 10.1016/j.redox.2024.103332] [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: 07/24/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Reactive Oxygen Species (ROS) refer to a variety of derivatives of molecular oxygen that play crucial roles in regulating a wide range of physiological and pathological processes. Excessive ROS levels can cause oxidative stress, leading to cellular damage and even cell demise. However, moderately elevated levels of ROS can mediate the oxidative post-translational modifications (oxPTMs) of redox-sensitive proteins, thereby affecting protein functions and regulating various cellular signaling pathways. Among the oxPTMs, ROS-induced reversible protein sulfenylation represents the initial form of cysteine oxidation for sensing redox signaling. In this review, we will summarize the discovery, chemical formation, and detection approaches of protein sulfenylation. In addition, we will highlight recent findings for the roles of protein sulfenylation in various diseases, including thrombotic disorders, diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer.
Collapse
Affiliation(s)
- Baoquan Mu
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Zeng
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Luo
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China.
| | - Kui Wang
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Sajjanar B, Aalam MT, Khan O, Dhara SK, Ghosh J, Gandham RK, Gupta PK, Chaudhuri P, Dutt T, Singh G, Mishra BP. Genome-wide DNA methylation profiles regulate distinct heat stress response in zebu (Bos indicus) and crossbred (Bos indicus × Bos taurus) cattle. Cell Stress Chaperones 2024; 29:603-614. [PMID: 38936463 PMCID: PMC11264184 DOI: 10.1016/j.cstres.2024.06.005] [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: 11/23/2023] [Revised: 05/22/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024] Open
Abstract
Epigenetic variations result from long-term adaptation to environmental factors. The Bos indicus (zebu) adapted to tropical conditions, whereas Bos taurus adapted to temperate conditions; hence native zebu cattle and its crossbred (B indicus × B taurus) show differences in responses to heat stress. The present study evaluated genome-wide DNA methylation profiles of these two breeds of cattle that may explain distinct heat stress responses. Physiological responses to heat stress and estimated values of Iberia heat tolerance coefficient and Benezra's coefficient of adaptability revealed better relative thermotolerance of Hariana compared to the Vrindavani cattle. Genome-wide DNA methylation patterns were different for Hariana and Vrindavani cattle. The comparison between breeds indicated the presence of 4599 significant differentially methylated CpGs with 756 hypermethylated and 3845 hypomethylated in Hariana compared to the Vrindavani cattle. Further, we found 79 genes that showed both differential methylation and differential expression that are involved in cellular stress response functions. Differential methylations in the microRNA coding sequences also revealed their functions in heat stress responses. Taken together, epigenetic differences represent the potential regulation of long-term adaptation of Hariana (B indicus) cattle to the tropical environment and relative thermotolerance.
Collapse
Affiliation(s)
- Basavaraj Sajjanar
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India; ICAR-Indian Veterinary Research Institute, Bengaluru Campus, Bengaluru, Karnataka, India.
| | - Mohd Tanzeel Aalam
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Owais Khan
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Sujoy K Dhara
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Jyotirmoy Ghosh
- ICAR-National Institute of Animal Nutrition and Physiology, Bengaluru, Karnataka, India
| | - Ravi Kumar Gandham
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Praveen K Gupta
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Pallab Chaudhuri
- ICAR-Indian Veterinary Research Institute, Bengaluru Campus, Bengaluru, Karnataka, India
| | - Triveni Dutt
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Gyanendra Singh
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India.
| | | |
Collapse
|
3
|
Camargo LL, Rios FJ, Montezano AC, Touyz RM. Reactive oxygen species in hypertension. Nat Rev Cardiol 2024:10.1038/s41569-024-01062-6. [PMID: 39048744 DOI: 10.1038/s41569-024-01062-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Hypertension is a leading risk factor for stroke, heart disease and chronic kidney disease. Multiple interacting factors and organ systems increase blood pressure and cause target-organ damage. Among the many molecular elements involved in the development of hypertension are reactive oxygen species (ROS), which influence cellular processes in systems that contribute to blood pressure elevation (such as the cardiovascular, renal, immune and central nervous systems, or the renin-angiotensin-aldosterone system). Dysregulated ROS production (oxidative stress) is a hallmark of hypertension in humans and experimental models. Of the many ROS-generating enzymes, NADPH oxidases are the most important in the development of hypertension. At the cellular level, ROS influence signalling pathways that define cell fate and function. Oxidative stress promotes aberrant redox signalling and cell injury, causing endothelial dysfunction, vascular damage, cardiovascular remodelling, inflammation and renal injury, which are all important in both the causes and consequences of hypertension. ROS scavengers reduce blood pressure in almost all experimental models of hypertension; however, clinical trials of antioxidants have yielded mixed results. In this Review, we highlight the latest advances in the understanding of the role and the clinical implications of ROS in hypertension. We focus on cellular sources of ROS, molecular mechanisms of oxidative stress and alterations in redox signalling in organ systems, and their contributions to hypertension.
Collapse
Affiliation(s)
- Livia L Camargo
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada.
| | - Francisco J Rios
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada.
- Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada.
- Department of Family Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
4
|
Lu HJ, Koju N, Sheng R. Mammalian integrated stress responses in stressed organelles and their functions. Acta Pharmacol Sin 2024; 45:1095-1114. [PMID: 38267546 PMCID: PMC11130345 DOI: 10.1038/s41401-023-01225-0] [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: 06/29/2023] [Accepted: 12/30/2023] [Indexed: 01/26/2024] Open
Abstract
The integrated stress response (ISR) triggered in response to various cellular stress enables mammalian cells to effectively cope with diverse stressful conditions while maintaining their normal functions. Four kinases (PERK, PKR, GCN2, and HRI) of ISR regulate ISR signaling and intracellular protein translation via mediating the phosphorylation of eukaryotic translation initiation factor 2 α (eIF2α) at Ser51. Early ISR creates an opportunity for cells to repair themselves and restore homeostasis. This effect, however, is reversed in the late stages of ISR. Currently, some studies have shown the non-negligible impact of ISR on diseases such as ischemic diseases, cognitive impairment, metabolic syndrome, cancer, vanishing white matter, etc. Hence, artificial regulation of ISR and its signaling with ISR modulators becomes a promising therapeutic strategy for relieving disease symptoms and improving clinical outcomes. Here, we provide an overview of the essential mechanisms of ISR and describe the ISR-related pathways in organelles including mitochondria, endoplasmic reticulum, Golgi apparatus, and lysosomes. Meanwhile, the regulatory effects of ISR modulators and their potential application in various diseases are also enumerated.
Collapse
Affiliation(s)
- Hao-Jun Lu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Nirmala Koju
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
5
|
Oh SH, Yook JM, Jung HY, Choi JY, Cho JH, Park SH, Kim CD, Kim YL, Lim JH. Autophagy caused by oxidative stress promotes TGF-β1-induced epithelial-to-mesenchymal transition in human peritoneal mesothelial cells. Cell Death Dis 2024; 15:365. [PMID: 38806451 PMCID: PMC11133371 DOI: 10.1038/s41419-024-06753-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: 11/24/2023] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
Epithelial-to-mesenchymal transition (EMT) is one of the main causes of peritoneal fibrosis. However, the pathophysiological mechanisms of EMT, specifically its relationship with autophagy, are still unknown. This study aimed to evaluate the role of autophagy in transforming growth factor-beta 1 (TGF-β1)-induced EMT in human peritoneal mesothelial cells (HPMCs). Primary cultured HPMCs were treated with TGF-β1 (2 and 5 ng/mL) and changes in autophagy markers and the relationship between autophagy and EMT were evaluated. We also identified changes in EMT- and autophagy-related signaling pathways after autophagy and NADPH oxidase 4 (NOX4) inhibition. TGF-β1 increased the generation of NOX4 and reactive oxygen species (ROS) in HPMCs, resulting in mitochondrial damage. Treatment with GKT137831 (20 μM), a NOX1/4 inhibitor, reduced ROS in the mitochondria of HPMC cells and reduced TGF-β1-induced mitochondrial damage. Additionally, the indirect inhibition of autophagy by GKT137831 (20 μM) downregulated TGF-β1-induced EMT, whereas direct inhibition of autophagy using 3-methyladenine (3-MA) (2 mM) or autophagy-related gene 5 (ATG5) gene silencing decreased the TGF-β1-induced EMT in HPMCs. The suppressor of mothers against decapentaplegic 2/3 (Smad2/3), autophagy-related phosphoinositide 3-kinase (PI3K) class III, and protein kinase B (Akt) pathways, and mitogen-activated protein kinase (MAPK) signaling pathways, such as extracellular signal-regulated kinase (ERK) and P38, were involved in TGF-β1-induced EMT. Autophagy and NOX4 inhibition suppressed the activation of these signaling pathways. Direct inhibition of autophagy and its indirect inhibition through the reduction of mitochondrial damage by upstream NOX4 inhibition reduced EMT in HPMCs. These results suggest that autophagy could serve as a therapeutic target for the prevention of peritoneal fibrosis in patients undergoing peritoneal dialysis.
Collapse
Affiliation(s)
- Se-Hyun Oh
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Ju-Min Yook
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hee-Yeon Jung
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Ji-Young Choi
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jang-Hee Cho
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Sun-Hee Park
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Chan-Duck Kim
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Yong-Lim Kim
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea.
| | - Jeong-Hoon Lim
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
6
|
Romashin D, Arzumanian V, Poverennaya E, Varshaver A, Luzgina N, Rusanov A. Evaluation of Cd-induced cytotoxicity in primary human keratinocytes. Hum Exp Toxicol 2024; 43:9603271231224458. [PMID: 38174414 DOI: 10.1177/09603271231224458] [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: 01/05/2024]
Abstract
An increasing number of studies have investigated the effects of Cd on human health. Cd-induced dermatotoxicity is an important field of research, but numerous studies have focused on the effects of Cd on the human skin. Moreover, most studies have been performed using HaCaT cells but not primary keratinocytes. In this study, we provide the results describing the cytotoxic effects of Cd exposure on primary human epidermal keratinocytes obtained from different donors. The subtoxic concentration of cadmium chloride was determined via MTT assay, and transcriptomic analysis of the cells exposed to this concentration (25 µM) was performed. As in HaCaT cells, Cd exposure resulted in increased ROS levels, cell cycle arrest, and induction of apoptosis. In addition, we report that exposure to Cd affects zinc and copper homeostasis, induces metallothionein expression, and activates various signaling pathways, including Nrf2, NF-kB, TRAIL, and PI3K. Cd induces the secretion of various cytokines (IL-1, IL-6, IL-10, and PGE2) and upregulates the expression of several cytokeratins, such as KRT6B, KRT6C, KRT16, and KRT17. The results provide a better understanding of the mechanisms of cadmium-induced cytotoxicity and its effect on human epidermal skin cells.
Collapse
|
7
|
Matsunaga S, Kohda A, Kamakura S, Hayase J, Miyano K, Shiose A, Sumimoto H. Hypoxia stabilizes the H 2 O 2 -producing oxidase Nox4 in cardiomyocytes via suppressing autophagy-related lysosomal degradation. Genes Cells 2024; 29:63-72. [PMID: 37985134 DOI: 10.1111/gtc.13085] [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: 10/23/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023]
Abstract
The hydrogen peroxide (H2 O2 )-producing NADPH oxidase Nox4, forming a heterodimer with p22phox , is expressed in a variety of cells including those in the heart to mediate adaptive responses to cellular stresses such as hypoxia. Since Nox4 is constitutively active, H2 O2 production is controlled by its protein abundance. Hypoxia-induced Nox4 expression is observed in various types of cells and generally thought to be regulated at the transcriptional level. Here we show that hypoxia upregulates the Nox4 protein level and Nox4-catalyzed H2 O2 production without increasing the Nox4 mRNA in rat H9c2 cardiomyocytes. In these cells, the Nox4 protein is stabilized under hypoxic conditions in a manner dependent on the presence of p22phox . Cell treatment with the proteasome inhibitor MG132 results in a marked decrease of the Nox4 protein under both normoxic and hypoxic conditions, indicating that the proteasome pathway does not play a major role in Nox4 degradation. The decrease is partially restored by the autophagy inhibitor 3-methyladenine. Furthermore, the Nox4 protein level is upregulated by the lysosome inhibitors bafilomycin A1 and chloroquine. Thus, in cardiomyocytes, Nox4 appears to be degraded via an autophagy-related pathway, and its suppression by hypoxia likely stabilizes Nox4, leading to upregulation of Nox4-catalyzed H2 O2 production.
Collapse
Affiliation(s)
- Shogo Matsunaga
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Akira Kohda
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Sachiko Kamakura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Junya Hayase
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kei Miyano
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Akira Shiose
- Department of Cardiovascular Surgery, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hideki Sumimoto
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
8
|
Camargo LL, Wang Y, Rios FJ, McBride M, Montezano AC, Touyz RM. Oxidative Stress and Endoplasmic Reticular Stress Interplay in the Vasculopathy of Hypertension. Can J Cardiol 2023; 39:1874-1887. [PMID: 37875177 DOI: 10.1016/j.cjca.2023.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 10/26/2023] Open
Abstract
Under physiologic conditions, reactive oxygen species (ROS) function as signalling molecules that control cell function. However, in pathologic conditions, increased generation of ROS triggers oxidative stress, which plays a role in vascular changes associated with hypertension, including endothelial dysfunction, vascular reactivity, and arterial remodelling (termed the vasculopathy of hypertension). The major source of ROS in the vascular system is NADPH oxidase (NOX). Increased NOX activity drives vascular oxidative stress in hypertension. Molecular mechanisms underlying vascular damage in hypertension include activation of redox-sensitive signalling pathways, post-translational modification of proteins, and oxidative damage of DNA and cytoplasmic proteins. In addition, oxidative stress leads to accumulation of proteins in the endoplasmic reticulum (ER) (termed ER stress), with consequent activation of the unfolded protein response (UPR). ER stress is emerging as a potential player in hypertension as abnormal protein folding in the ER leads to oxidative stress and dysregulated activation of the UPR promotes inflammation and injury in vascular and cardiac cells. In addition, the ER engages in crosstalk with exogenous sources of ROS, such as mitochondria and NOX, which can amplify redox processes. Here we provide an update of the role of ROS and NOX in hypertension and discuss novel concepts on the interplay between oxidative stress and ER stress.
Collapse
Affiliation(s)
- Livia L Camargo
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| | - Yu Wang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Francisco J Rios
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Martin McBride
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; McGill University, Department of Medicine and Department of Family Medicine, Montréal, Québec, Canada.
| |
Collapse
|
9
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
10
|
Huang C, Han X, Yang L, Song W, Zhang H, Zhu X, Huang G, Xu J. Exosomal miR-129 and miR-342 derived from intermittent hypoxia-stimulated vascular smooth muscle cells inhibit the eIF2α/ATF4 axis from preventing calcified aortic valvular disease. J Cell Commun Signal 2023:10.1007/s12079-023-00785-4. [PMID: 37812275 DOI: 10.1007/s12079-023-00785-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
This study aims to elucidate the role of miR-129/miR-342 loaded in exosomes derived from vascular smooth muscle cells (VSMCs) stimulated by intermittent hypoxia in calcified aortic valvular disease (CAVD). Bioinformatics analysis was conducted to identify differentially expressed miRs in VSMCs-derived exosomes and CAVD samples, and their potential target genes were predicted. VSMCs were exposed to intermittent hypoxia to induce stimulation, followed by isolation of exosomes. Valvular interstitial cells (VICs) were cultured in vitro to investigate the impact of miR-129/miR-342 on VICs' osteogenic differentiation and aortic valve calcification with eIF2α. A CAVD mouse model was established using ApoE knockout mice for in vivo validation. In CAVD samples, miR-129 and miR-342 were downregulated, while eIF2α and ATF4 were upregulated. miR-129 and miR-342 exhibited inhibitory effects on eIF2α through targeted regulation. Exosomes released from intermittently hypoxia-stimulated VSMCs contained miR-129 and miR-342. Overexpression of miR-129 and miR-342, or silencing ATF4, suppressed VICs' osteogenic differentiation and aortic valve calcification, which could be rescued by overexpressed eIF2α. Collectively, intermittent hypoxia stimulation of VSMCs leads to the secretion of exosomes that activate the miR-129/miR-342 dual pathway, thereby inhibiting the eIF2α/ATF4 axis and attenuating VICs' osteogenic differentiation and CAVD progression.
Collapse
Affiliation(s)
- Chen Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Xu Han
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Linjie Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Wei Song
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Hualu Zhang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaohua Zhu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Gongcheng Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China
| | - Jing Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Erqi Distrcit, Zhengzhou, 450000, Henan Province, China.
| |
Collapse
|
11
|
Li J, Wang L, Wang B, Zhang Z, Jiang L, Qin Z, Zhao Y, Su B. NOX4 is a potential therapeutic target in septic acute kidney injury by inhibiting mitochondrial dysfunction and inflammation. Theranostics 2023; 13:2863-2878. [PMID: 37284448 PMCID: PMC10240817 DOI: 10.7150/thno.81240] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 04/25/2023] [Indexed: 06/08/2023] Open
Abstract
Rationale: Sepsis is a severe clinical syndrome featured through organ dysfunction due to infection, while the accompanying acute kidney injury (AKI) is linked to significant incidence of morbidity as well as mortality. Recently, emerging evidence has revealed that nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4 (NOX4) is implicated in various renal diseases, while its role and modulation in septic acute kidney injury (S-AKI) remains largely unknown. Methods: In vivo, S-AKI in wild-type and renal tubular epithelial cell (RTEC)-specific NOX4 knockout mice was induced by lipopolysaccharides (LPS) injection or cecal ligation and puncture (CLP). In vitro, TCMK-1 (mouse kidney tubular epithelium cell line) cells were treated with LPS. Serum and supernatant biochemical, mitochondrial dysfunctional, inflammatory and apoptotic parameters were measured and compared across groups. The activation of reactive oxygen species (ROS) and NF-κB signaling was also assessed. Results: NOX4 was predominantly upregulated in RTECs of S-AKI mouse model induced by LPS/CLP and cultured TCMK-1 cells exposed to LPS. RTEC-specific deletion of NOX4 or pharmacological inhibition of NOX4 by GKT137831 both alleviated LPS/CLP-injured renal function and pathology in mice. Furthermore, NOX4 inhibition alleviated mitochondrial dysfunction supported by ultrastructural damage, reduction of ATP production and mitochondrial dynamics imbalance, together with inflammation and apoptosis in kidney injured by LPS/CLP and TCMK-1 cells injured by LPS, while NOX4 overexpression aggravated the above-mentioned indices in TCMK-1 cells with LPS stimulation. Mechanism-wise, the raised NOX4 in RTECs may induce ROS and NF-κB signaling activation in S-AKI. Conclusions: Collectively, genetic or pharmacological inhibition of NOX4 protects from S-AKI by reducing generation of ROS and activation of NF-κB signal, which suppress mitochondrial dysfunction, inflammation together with apoptosis. NOX4 may act as a novel target for the S-AKI therapy.
Collapse
Affiliation(s)
- Jiameng Li
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liya Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhuyun Zhang
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Luojia Jiang
- Department of Nephrology, Jiujiang No. 1 People's Hospital, Jiujiang 332000, China
| | - Zheng Qin
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuliang Zhao
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Baihai Su
- Department of Nephrology, Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
12
|
Melatonin Mitigates iNOS-Related Effects of HEMA and Camphorquinone in Human Dental Pulp Cells: Relevance for Postoperative Sensitivity Mechanism in Type 2 Diabetes. Int J Mol Sci 2023; 24:ijms24032562. [PMID: 36768884 PMCID: PMC9916734 DOI: 10.3390/ijms24032562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/09/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
High elution and diffusion of 2-hydroxylethyl methacrylate (HEMA) and camphorquinone (CQ) through dentinal tubules may induce pulp injury and postoperative sensitivity. We aimed to investigate the melatonin protective effect in HEMA- and CQ-treated human dental pulp cells (hDPCs) as well as its relevance in a mechanism for postoperative sensitivity in diabetic patients. hDPCs were exposed to HEMA (5 mM) and/or CQ (1 mM) in the absence and presence of melatonin (MEL) (0.1 mM and 1 mM). Heme oxygenase-1 (HMOX1), NADPH oxidase-4 (NOX4), BCL-2-associated X-protein (BAX), B-cell lymphoma-2 (BCL-2) and caspase-3 (CASP3) gene expression levels, and superoxide dismutase (SOD) activity were measured in hDPCs while inducible nitric oxide synthase (iNOS) and melatonin protein expression were measured in human dental pulp as well, by RT-PCR, by ELISA, and spectrophotometrically. Bioinformatic analyses were performed by using the ShinyGO (v.0.75) application. Type 2 diabetic patients showed a higher incidence of postoperative sensitivity and lower melatonin and higher iNOS content in dental pulp tissue compared with non-diabetic patients. Melatonin, when co-added in hDPC culture, reverses HEMA and CQ cytotoxic effects via anti-apoptotic and anti-inflammatory/antioxidant iNOS-related effects. Enrichment analyses showed that genes/proteins, altered by HEMA and CQ and normalized by melatonin, are the most prominently overrepresented in type 2 diabetes mellitus pathways and that they share subcellular localization in different oligomeric protein complexes consisting of anti- and pro-apoptotic regulators. This is the first evidence of the ability of melatonin to counteract iNOS-mediated inflammatory and stress effects in HEMA- and CQ-treated hDPCs, which could be of significance for the modulation of presently observed immediate postoperative sensitivity after composite restoration in type 2 diabetic patients.
Collapse
|
13
|
Nabeebaccus AA, Reumiller CM, Shen J, Zoccarato A, Santos CXC, Shah AM. The regulation of cardiac intermediary metabolism by NADPH oxidases. Cardiovasc Res 2023; 118:3305-3319. [PMID: 35325070 PMCID: PMC9847558 DOI: 10.1093/cvr/cvac030] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/24/2021] [Accepted: 01/18/2022] [Indexed: 01/25/2023] Open
Abstract
NADPH oxidases (NOXs), enzymes whose primary function is to generate reactive oxygen species, are important regulators of the heart's physiological function and response to pathological insults. The role of NOX-driven redox signalling in pathophysiological myocardial remodelling, including processes such as interstitial fibrosis, contractile dysfunction, cellular hypertrophy, and cell survival, is well recognized. While the NOX2 isoform promotes many detrimental effects, the NOX4 isoform has attracted considerable attention as a driver of adaptive stress responses both during pathology and under physiological states such as exercise. Recent studies have begun to define some of the NOX4-modulated mechanisms that may underlie these adaptive responses. In particular, novel functions of NOX4 in driving cellular metabolic changes have emerged. Alterations in cellular metabolism are a recognized hallmark of the heart's response to physiological and pathological stresses. In this review, we highlight the emerging roles of NOX enzymes as important modulators of cellular intermediary metabolism in the heart, linking stress responses not only to myocardial energetics but also other functions. The novel interplay of NOX-modulated redox signalling pathways and intermediary metabolism in the heart is unravelling a new aspect of the fascinating biology of these enzymes which will inform a better understanding of how they drive adaptive responses. We also discuss the implications of these new findings for therapeutic approaches that target metabolism in cardiac disease.
Collapse
Affiliation(s)
- Adam A Nabeebaccus
- School of Cardiovascular Medicine and Sciences, King’s College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Christina M Reumiller
- School of Cardiovascular Medicine and Sciences, King’s College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Jie Shen
- School of Cardiovascular Medicine and Sciences, King’s College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Anna Zoccarato
- School of Cardiovascular Medicine and Sciences, King’s College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Celio X C Santos
- School of Cardiovascular Medicine and Sciences, King’s College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, King’s College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
14
|
Huang Q, Chen Y, Zhang Z, Xue Z, Hua Z, Luo X, Li Y, Lu C, Lu A, Liu Y. The endoplasmic reticulum participated in drug metabolic toxicity. Cell Biol Toxicol 2022; 38:945-961. [PMID: 35040016 DOI: 10.1007/s10565-021-09689-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/07/2021] [Indexed: 01/25/2023]
Abstract
Covalent binding of reactive metabolites formed by drug metabolic activation with biological macromolecules is considered to be an important mechanism of drug metabolic toxicity. Recent studies indicate that the endoplasmic reticulum (ER) could play an important role in drug toxicity by participating in the metabolic activation of drugs and could be a primarily attacked target by reactive metabolites. In this article, we summarize the generation and mechanism of reactive metabolites in ER stress and their associated cell death and inflammatory cascade, as well as the systematic modulation of unfolded protein response (UPR)-mediated adaptive pathways.
Collapse
Affiliation(s)
- Qingcai Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Youwen Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhengjia Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zeyu Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
15
|
Dong Q, Zhang X, Zhang M. Long Noncoding RNA-5657 Inhibits the Proliferation of Pancreatic Cells and Insulin Secretion in Gestational Diabetes by Inhibiting the NADPH Oxidase 4 (NOX4). J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Multiple comorbidities are observed during pregnancy and there are many challenges for pregnant women with a previous history of diabetes. Pancreatic β-cell dysfunction is associated with gestational diabetes mellitus (GDM). This study intends to explore LncRNA-5657’s
effect on proapoptotic cell proliferation and insulin secretion in gestational diabetes. Real-time quantitative fluorescence PCR measured LncRNA-5657 expression in GDM patient placental tissues along with analysis of cell activity, proliferation and insulin secretion by MTT assay, BrdU staining
and ELISA. The relation between LncRNA-5657 and NOX4 was assessed by dual luciferase reporter assay. LncRNA-5657 was overexpressed in placental tissue of GDM patients compared to normal pregnant women. LncRNA-5657 overexpression promoted cell proliferation and insulin secretion, which was
reversed by LncRNA-5657 downregulation. In addition, LncRNA-5657 negatively regulated NOX4 expression. Overexpression of NOX4 abolished the effect of LncRNA-5657 mimics on pancreatic β-cells, and knockdown of LncRNA-5657 promoted insulin secretion by upregulating NOX4 expression.
In conclusion, LncRNA-5657 inhibits insulin secretion and β-cell proliferation by targeting NOX4, indicating that LncRNA-5657 might be a novel target for the treatment of GDM.
Collapse
Affiliation(s)
- Qingqing Dong
- Department of Endocrinology, Hangzhou Fuyang Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, China
| | - Xiaohong Zhang
- Department of Endocrinology, Hangzhou Fuyang Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, China
| | - Min Zhang
- Department of Nursing of Traditional Chinese Medicine, Hangzhou Fuyang Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, China
| |
Collapse
|
16
|
Guo CL. Self-Sustained Regulation or Self-Perpetuating Dysregulation: ROS-dependent HIF-YAP-Notch Signaling as a Double-Edged Sword on Stem Cell Physiology and Tumorigenesis. Front Cell Dev Biol 2022; 10:862791. [PMID: 35774228 PMCID: PMC9237464 DOI: 10.3389/fcell.2022.862791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/29/2022] [Indexed: 12/19/2022] Open
Abstract
Organ development, homeostasis, and repair often rely on bidirectional, self-organized cell-niche interactions, through which cells select cell fate, such as stem cell self-renewal and differentiation. The niche contains multiplexed chemical and mechanical factors. How cells interpret niche structural information such as the 3D topology of organs and integrate with multiplexed mechano-chemical signals is an open and active research field. Among all the niche factors, reactive oxygen species (ROS) have recently gained growing interest. Once considered harmful, ROS are now recognized as an important niche factor in the regulation of tissue mechanics and topology through, for example, the HIF-YAP-Notch signaling pathways. These pathways are not only involved in the regulation of stem cell physiology but also associated with inflammation, neurological disorder, aging, tumorigenesis, and the regulation of the immune checkpoint molecule PD-L1. Positive feedback circuits have been identified in the interplay of ROS and HIF-YAP-Notch signaling, leading to the possibility that under aberrant conditions, self-organized, ROS-dependent physiological regulations can be switched to self-perpetuating dysregulation, making ROS a double-edged sword at the interface of stem cell physiology and tumorigenesis. In this review, we discuss the recent findings on how ROS and tissue mechanics affect YAP-HIF-Notch-PD-L1 signaling, hoping that the knowledge can be used to design strategies for stem cell-based and ROS-targeting therapy and tissue engineering.
Collapse
Affiliation(s)
- Chin-Lin Guo
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
17
|
Greenberg HZE, Zhao G, Shah AM, Zhang M. Role of oxidative stress in calcific aortic valve disease and its therapeutic implications. Cardiovasc Res 2022; 118:1433-1451. [PMID: 33881501 PMCID: PMC9074995 DOI: 10.1093/cvr/cvab142] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the end result of active cellular processes that lead to the progressive fibrosis and calcification of aortic valve leaflets. In western populations, CAVD is a significant cause of cardiovascular morbidity and mortality, and in the absence of effective drugs, it will likely represent an increasing disease burden as populations age. As there are currently no pharmacological therapies available for preventing, treating, or slowing the development of CAVD, understanding the mechanisms underlying the initiation and progression of the disease is important for identifying novel therapeutic targets. Recent evidence has emerged of an important causative role for reactive oxygen species (ROS)-mediated oxidative stress in the pathophysiology of CAVD, inducing the differentiation of valve interstitial cells into myofibroblasts and then osteoblasts. In this review, we focus on the roles and sources of ROS driving CAVD and consider their potential as novel therapeutic targets for this debilitating condition.
Collapse
Affiliation(s)
- Harry Z E Greenberg
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Guoan Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Henan, China
| | - Ajay M Shah
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Min Zhang
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
18
|
An Overview of the Molecular Mechanisms Associated with Myocardial Ischemic Injury: State of the Art and Translational Perspectives. Cells 2022; 11:cells11071165. [PMID: 35406729 PMCID: PMC8998015 DOI: 10.3390/cells11071165] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease is the leading cause of death in western countries. Among cardiovascular diseases, myocardial infarction represents a life-threatening condition predisposing to the development of heart failure. In recent decades, much effort has been invested in studying the molecular mechanisms underlying the development and progression of ischemia/reperfusion (I/R) injury and post-ischemic cardiac remodeling. These mechanisms include metabolic alterations, ROS overproduction, inflammation, autophagy deregulation and mitochondrial dysfunction. This review article discusses the most recent evidence regarding the molecular basis of myocardial ischemic injury and the new potential therapeutic interventions for boosting cardioprotection and attenuating cardiac remodeling.
Collapse
|
19
|
Müller N, Warwick T, Noack K, Malacarne PF, Cooper AJL, Weissmann N, Schröder K, Brandes RP, Rezende F. Reactive Oxygen Species Differentially Modulate the Metabolic and Transcriptomic Response of Endothelial Cells. Antioxidants (Basel) 2022; 11:antiox11020434. [PMID: 35204316 PMCID: PMC8869421 DOI: 10.3390/antiox11020434] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Reactive oxygen species (ROS) are important mediators of both physiological and pathophysiological signal transduction in the cardiovascular system. The effects of ROS on cellular processes depend on the concentration, localization, and duration of exposure. Cellular stress response mechanisms have evolved to mitigate the negative effects of acute oxidative stress. In this study, we investigate the short-term and long-term metabolic and transcriptomic response of human umbilical vein endothelial cells (HUVEC) to different types and concentrations of ROS. To generate intracellular H2O2, we utilized a lentiviral chemogenetic approach for overexpression of human D-amino acid oxidase (DAO). DAO converts D-amino acids into their corresponding imino acids and H2O2. HUVEC stably overexpressing DAO (DAO-HUVEC) were exposed to D-alanine (3 mM), exogenous H2O2 (10 µM or 300 µM), or menadione (5 µM) for various timepoints and subjected to global untargeted metabolomics (LC-MS/MS) and RNAseq by MACE (Massive analysis of cDNA ends). A total of 300 µM H2O2 led to pronounced changes on both the metabolic and transcriptomic level. In particular, metabolites linked to redox homeostasis, energy-generating pathways, and nucleotide metabolism were significantly altered. Furthermore, 300 µM H2O2 affected genes related to the p53 pathway and cell cycle. In comparison, the effects of menadione and DAO-derived H2O2 mainly occurred at gene expression level. Collectively, all types of ROS led to subtle changes in the expression of ribosomal genes. Our results show that different types and concentration of ROS lead to a different metabolic and transcriptomic response in endothelial cells.
Collapse
Affiliation(s)
- Niklas Müller
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany; (N.M.); (T.W.); (K.N.); (P.F.M.); (K.S.); (R.P.B.)
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, 60590 Frankfurt, Germany
| | - Timothy Warwick
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany; (N.M.); (T.W.); (K.N.); (P.F.M.); (K.S.); (R.P.B.)
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, 60590 Frankfurt, Germany
| | - Kurt Noack
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany; (N.M.); (T.W.); (K.N.); (P.F.M.); (K.S.); (R.P.B.)
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, 60590 Frankfurt, Germany
| | - Pedro Felipe Malacarne
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany; (N.M.); (T.W.); (K.N.); (P.F.M.); (K.S.); (R.P.B.)
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, 60590 Frankfurt, Germany
| | - Arthur J. L. Cooper
- Department of Biochemistry and Molecular Biology, New York Medical College, 15 Dana Road, Valhalla, NY 10595, USA;
| | - Norbert Weissmann
- Justus 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, 35390 Giessen, Germany;
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany; (N.M.); (T.W.); (K.N.); (P.F.M.); (K.S.); (R.P.B.)
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, 60590 Frankfurt, Germany
| | - Ralf P. Brandes
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany; (N.M.); (T.W.); (K.N.); (P.F.M.); (K.S.); (R.P.B.)
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, 60590 Frankfurt, Germany
| | - Flávia Rezende
- Institute for Cardiovascular Physiology, Goethe University, Theodor-Stern Kai 7, 60590 Frankfurt, Germany; (N.M.); (T.W.); (K.N.); (P.F.M.); (K.S.); (R.P.B.)
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, 60590 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-6301-85321; Fax: +49-69-6301-7668
| |
Collapse
|
20
|
Fang J, Sheng R, Qin ZH. NADPH Oxidases in the Central Nervous System: Regional and Cellular Localization and the Possible Link to Brain Diseases. Antioxid Redox Signal 2021; 35:951-973. [PMID: 34293949 DOI: 10.1089/ars.2021.0040] [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: 01/03/2023]
Abstract
Significance: The significant role of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) in signal transduction is mediated by the production of reactive oxygen species (ROS), especially in the central nervous system (CNS). The pathogenesis of some neurologic and psychiatric diseases is regulated by ROS, acting as a second messenger or pathogen. Recent Advances: In the CNS, the involvement of Nox-derived ROS has been implicated in the regulation of multiple signals, including cell survival/apoptosis, neuroinflammation, migration, differentiation, proliferation, and synaptic plasticity, as well as the integrity of the blood/brain barrier. In these processes, the intracellular signals mediated by the members of the Nox family vary among different tissues. The present review illuminates the regions and cellular, subcellular localization of Nox isoforms in the brain, the signal transduction, and the role of NOX enzymes in pathophysiology, respectively. Critical Issues: Different signal transduction cascades are coupled to ROS derived from various Nox homologues with varying degrees. Therefore, a critical issue worth noting is the varied role of the homologues of NOX enzymes in different signaling pathways and also they mediate different phenotypes in the diverse pathophysiological condition. This substantiates the effectiveness of selective Nox inhibitors in the CNS. Future Directions: Further investigation to elucidate the role of various homologues of NOX enzymes in acute and chronic brain diseases and signaling mechanisms, and the development of more specific NOX inhibitors for the treatment of CNS disease are urgently needed. Antioxid. Redox Signal. 35, 951-973.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| |
Collapse
|
21
|
Viswambharan H, Yuldasheva NY, Imrie H, Bridge K, Haywood NJ, Skromna A, Hemmings KE, Clark ER, Gatenby VK, Cordell P, Simmons KJ, Makava N, Abudushalamu Y, Endesh N, Brown J, Walker AMN, Futers ST, Porter KE, Cubbon RM, Naseem K, Shah AM, Beech DJ, Wheatcroft SB, Kearney MT, Sukumar P. Novel Paracrine Action of Endothelium Enhances Glucose Uptake in Muscle and Fat. Circ Res 2021; 129:720-734. [PMID: 34420367 PMCID: PMC8448413 DOI: 10.1161/circresaha.121.319517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hema Viswambharan
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Nadira Y Yuldasheva
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Helen Imrie
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Katherine Bridge
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Natalie J Haywood
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Anna Skromna
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Karen E Hemmings
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Emily R Clark
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - V Kate Gatenby
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Paul Cordell
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Katie J Simmons
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Natallia Makava
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Yilizila Abudushalamu
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Naima Endesh
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Jane Brown
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Andrew M N Walker
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Simon T Futers
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Karen E Porter
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Richard M Cubbon
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Khalid Naseem
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Ajay M Shah
- British Heart Foundation Centre of Research Excellence, King's College London (A.M.S.)
| | - David J Beech
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Stephen B Wheatcroft
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Piruthivi Sukumar
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| |
Collapse
|
22
|
Zoccarato A, Nabeebaccus AA, Oexner RR, Santos CXC, Shah AM. The nexus between redox state and intermediary metabolism. FEBS J 2021; 289:5440-5462. [PMID: 34496138 DOI: 10.1111/febs.16191] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/20/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
Reactive oxygen species (ROS) are not just a by-product of cellular metabolic processes but act as signalling molecules that regulate both physiological and pathophysiological processes. A close connection exists in cells between redox homeostasis and cellular metabolism. In this review, we describe how intracellular redox state and glycolytic intermediary metabolism are closely coupled. On the one hand, ROS signalling can control glycolytic intermediary metabolism by direct regulation of the activity of key metabolic enzymes and indirect regulation via redox-sensitive transcription factors. On the other hand, metabolic adaptation and reprogramming in response to physiological or pathological stimuli regulate intracellular redox balance, through mechanisms such as the generation of reducing equivalents. We also discuss the impact of these intermediary metabolism-redox circuits in physiological and disease settings across different tissues. A better understanding of the mechanisms regulating these intermediary metabolism-redox circuits will be crucial to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Anna Zoccarato
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Adam A Nabeebaccus
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Rafael R Oexner
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Celio X C Santos
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| |
Collapse
|
23
|
Sinenko SA, Starkova TY, Kuzmin AA, Tomilin AN. Physiological Signaling Functions of Reactive Oxygen Species in Stem Cells: From Flies to Man. Front Cell Dev Biol 2021; 9:714370. [PMID: 34422833 PMCID: PMC8377544 DOI: 10.3389/fcell.2021.714370] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS), superoxide anion and hydrogen peroxide, are generated as byproducts of oxidative phosphorylation in the mitochondria or via cell signaling-induced NADPH oxidases in the cytosol. In the recent two decades, a plethora of studies established that elevated ROS levels generated by oxidative eustress are crucial physiological mediators of many cellular and developmental processes. In this review, we discuss the mechanisms of ROS generation and regulation, current understanding of ROS functions in the maintenance of adult and embryonic stem cells, as well as in the process of cell reprogramming to a pluripotent state. Recently discovered cell-non-autonomous ROS functions mediated by growth factors are crucial for controlling cell differentiation and cellular immune response in Drosophila. Importantly, many physiological functions of ROS discovered in Drosophila may allow for deciphering and understanding analogous processes in human, which could potentially lead to the development of novel therapeutic approaches in ROS-associated diseases treatment.
Collapse
Affiliation(s)
- Sergey A Sinenko
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | | | - Andrey A Kuzmin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Alexey N Tomilin
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
24
|
Weissman D, Maack C. Redox signaling in heart failure and therapeutic implications. Free Radic Biol Med 2021; 171:345-364. [PMID: 34019933 DOI: 10.1016/j.freeradbiomed.2021.05.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/17/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022]
Abstract
Heart failure is a growing health burden worldwide characterized by alterations in excitation-contraction coupling, cardiac energetic deficit and oxidative stress. While current treatments are mostly limited to antagonization of neuroendocrine activation, more recent data suggest that also targeting metabolism may provide substantial prognostic benefit. However, although in a broad spectrum of preclinical models, oxidative stress plays a causal role for the development and progression of heart failure, no treatment that targets reactive oxygen species (ROS) directly has entered the clinical arena yet. In the heart, ROS derive from various sources, such as NADPH oxidases, xanthine oxidase, uncoupled nitric oxide synthase and mitochondria. While mitochondria are the primary source of ROS in the heart, communication between different ROS sources may be relevant for physiological signalling events as well as pathologically elevated ROS that deteriorate excitation-contraction coupling, induce hypertrophy and/or trigger cell death. Here, we review the sources of ROS in the heart, the modes of pathological activation of ROS formation as well as therapeutic approaches that may target ROS specifically in mitochondria.
Collapse
Affiliation(s)
- David Weissman
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany; Department of Internal Medicine 1, University Clinic Würzburg, Würzburg, Germany.
| |
Collapse
|
25
|
Nox4 Maintains Blood Pressure during Low Sodium Diet. Antioxidants (Basel) 2021; 10:antiox10071103. [PMID: 34356336 PMCID: PMC8301203 DOI: 10.3390/antiox10071103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 01/26/2023] Open
Abstract
The NADPH oxidase Nox4 is a hydrogen peroxide (H2O2)-producing enzyme, with the highest expression in the kidney. As the kidney is involved in volume and blood pressure control through sodium handling, we set out to determine the impact of a low sodium diet on these parameters in WT and Nox4-/- mice. Nox4 expression in the murine kidney was restricted to the proximal tubule. Nevertheless, low-sodium-induced weight loss and sodium sparing function was similar in WT and Nox4-/- mice, disputing an important function of renal Nox4 in sodium handling. In contrast, a low sodium diet resulted in a reduction in systolic blood pressure in Nox4-/- as compared to WT mice. This was associated with a selectively lower pressure to heart-rate ratio, as well as heart to body weight ratio. In general, a low sodium diet leads to activation of sympathetic tone and the renin angiotensin system, which subsequently increases peripheral resistance. Our observations suggest that the control by this system is attenuated in Nox4-/- mice, resulting in lower blood pressure in response to low sodium.
Collapse
|
26
|
Manuelli V, Pecorari C, Filomeni G, Zito E. Regulation of redox signaling in HIF-1-dependent tumor angiogenesis. FEBS J 2021; 289:5413-5425. [PMID: 34228878 DOI: 10.1111/febs.16110] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/24/2021] [Accepted: 07/05/2021] [Indexed: 12/11/2022]
Abstract
Angiogenesis is the process of blood vessel growth. The angiogenic switch consists of new blood vessel formation that, in carcinogenesis, can lead to the transition from a harmless cluster of dormant cells to a large tumorigenic mass with metastatic potential. Hypoxia, that is, the scarcity of oxygen, is a hallmark of solid tumors to which they adapt by activating hypoxia-inducible factor-1 (HIF-1), a transcription factor triggering de novo angiogenesis. HIF-1 and the angiogenic molecules that are expressed upon its activation are modulated by redox status. Modulations of the redox environment can influence the angiogenesis signaling at different levels, thereby impinging on the angiogenic switch. This review provides a molecular overview of the redox-sensitive steps in angiogenic signaling, the main molecular players involved, and their crosstalk with the unfolded protein response. New classes of inhibitors of these modulators which might act as antiangiogenic drugs in cancer are also discussed.
Collapse
Affiliation(s)
- Valeria Manuelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Chiara Pecorari
- Redox Biology Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Giuseppe Filomeni
- Redox Biology Group, Danish Cancer Society Research Center, Copenhagen, Denmark.,Center for Healthy Aging, Copenhagen University, Denmark.,Department of Biology, Tor Vergata University, Rome, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.,Department of Biomolecular Sciences, University of Urbino Carlo Bo, Italy
| |
Collapse
|
27
|
Brewer AC. Physiological interrelationships between NADPH oxidases and chromatin remodelling. Free Radic Biol Med 2021; 170:109-115. [PMID: 33556461 DOI: 10.1016/j.freeradbiomed.2021.01.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/08/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
The epigenetic landscape describes the chromatin structure of the eukaryotic genome and is therefore the major determinant of gene transcription and hence cellular phenotype. The molecular processes which act to shape the epigenetic landscape through cellular differentiation are thus central to cellular determination and specification. In addition, cellular adaptation to (patho)-physiological stress requires dynamic and reversible chromatin remodelling. It is becoming clear that redox-dependent molecular mechanisms are important determinants of this epigenetic regulation. NADPH oxidases generate reactive oxygen species (ROS) to activate redox-dependent signalling pathways in response to extracellular and intracellular environmental cues. This mini review aims to summarise the current knowledge of the role of NADPH oxidases in redox-dependent chromatin remodelling, and how epigenetic changes might feedback and impact upon the transcriptional expression of these ROS-producing enzymes themselves. The potential physiological significance of this relationship in the control of cellular differentiation and homeostasis by Nox4, specifically, is discussed.
Collapse
Affiliation(s)
- Alison C Brewer
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK.
| |
Collapse
|
28
|
Huang TC, Luo L, Jiang SH, Chen C, He HY, Liang CF, Li WS, Wang H, Zhu L, Wang K, Guo Y. Targeting integrated stress response regulates microglial M1/M2 polarization and attenuates neuroinflammation following surgical brain injury in rat. Cell Signal 2021; 85:110048. [PMID: 34015470 DOI: 10.1016/j.cellsig.2021.110048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/01/2021] [Accepted: 05/16/2021] [Indexed: 12/24/2022]
Abstract
Integrated stress response (ISR) contributes to various neuropathological processes and acting as a therapy target in CNS injuries. However, the fundamental role of ISR in regulating microglial polarization remains largely unknown. Currently no proper pharmacological approaches to reverse microglia-driven neuroinflammation in surgical brain injury (SBI) have been reported. Here we found that inhibition of the crucial ISR effector, activating transcription factor 4 (ATF4), using the RNA interference suppressed the lipopolysaccharide (LPS)-stimulated microglial M1 polarization in vitro. Interestingly, counteracting ISR with a small-molecule ISR inhibitor (ISRIB) resulted in a significant microglial M1 towards M2 phenotype switching after LPS treatment. The potential underlying mechanisms may related to downregulate the intracellular NADPH oxidase 4 (NOX4) expression under the neuroinflammatory microenvironment. Notably, ISRIB ameliorated the infiltration of microglia and improved the neurobehavioral outcomes in the SBI rat model. Overall, our findings suggest that targeting ISR exerts a novel anti-inflammatory effect on microglia via regulating M1/M2 phenotype and may represent a potential therapeutic target to overcome neuroinflammation following SBI.
Collapse
Affiliation(s)
- Teng-Chao Huang
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China; East China Institute of Digital Medical Engineering, Shangrao 334000, PR China
| | - Lun Luo
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Shi-Hai Jiang
- Department of Joint Replacement and Trauma Surgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China; Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany
| | - Chuan Chen
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Hai-Yong He
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Chao-Feng Liang
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Wen-Sheng Li
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Hui Wang
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Lei Zhu
- Department of Burns, Plastic & Reconstructive Surgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China
| | - Kun Wang
- Department of Joint Replacement and Trauma Surgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China.
| | - Ying Guo
- Department of Neurosurgery, Third Affiliated Hospital of Sun Yat-sen University, Canton 510630, PR China.
| |
Collapse
|
29
|
Regulation of Cardiac PKA Signaling by cAMP and Oxidants. Antioxidants (Basel) 2021; 10:antiox10050663. [PMID: 33923287 PMCID: PMC8146537 DOI: 10.3390/antiox10050663] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/31/2022] Open
Abstract
Pathologies, such as cancer, inflammatory and cardiac diseases are commonly associated with long-term increased production and release of reactive oxygen species referred to as oxidative stress. Thereby, protein oxidation conveys protein dysfunction and contributes to disease progression. Importantly, trials to scavenge oxidants by systemic antioxidant therapy failed. This observation supports the notion that oxidants are indispensable physiological signaling molecules that induce oxidative post-translational modifications in target proteins. In cardiac myocytes, the main driver of cardiac contractility is the activation of the β-adrenoceptor-signaling cascade leading to increased cellular cAMP production and activation of its main effector, the cAMP-dependent protein kinase (PKA). PKA-mediated phosphorylation of substrate proteins that are involved in excitation-contraction coupling are responsible for the observed positive inotropic and lusitropic effects. PKA-actions are counteracted by cellular protein phosphatases (PP) that dephosphorylate substrate proteins and thus allow the termination of PKA-signaling. Both, kinase and phosphatase are redox-sensitive and susceptible to oxidation on critical cysteine residues. Thereby, oxidation of the regulatory PKA and PP subunits is considered to regulate subcellular kinase and phosphatase localization, while intradisulfide formation of the catalytic subunits negatively impacts on catalytic activity with direct consequences on substrate (de)phosphorylation and cardiac contractile function. This review article attempts to incorporate the current perception of the functionally relevant regulation of cardiac contractility by classical cAMP-dependent signaling with the contribution of oxidant modification.
Collapse
|
30
|
Interactions of zinc- and redox-signaling pathways. Redox Biol 2021; 41:101916. [PMID: 33662875 PMCID: PMC7937829 DOI: 10.1016/j.redox.2021.101916] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
Zinc and cellular oxidants such as reactive oxygen species (ROS) each participate in a multitude of physiological functions. There is considerable overlap between the affected events, including signal transduction. While there is no obvious direct connection between zinc and ROS, mainly because the bivalent cation zinc does not change its oxidation state in biological systems, these are linked by their interaction with sulfur, forming the remarkable triad of zinc, ROS, and protein thiols. First, zinc binds to reduced thiols and can be released upon oxidation. Thereby, redox signals are translated into changes in the free zinc concentration, which can act as zinc signals. Second, zinc affects oxidation of thiols in several ways, directly as well as indirectly. A protein incorporating many of these interactions is metallothionein (MT), which is rich in cysteine and capable of binding up to seven zinc ions in its fully reduced state. Zinc binding is diminished after (partial) oxidation, while thiols show increased reactivity in the absence of bound metal ions. Adding still more complexity, the MT promoter is controlled by zinc (via metal regulatory transcription factor 1 (MTF-1)) as well as redox (via nuclear factor erythroid 2-related factor 2 (NRF2)). Many signaling cascades that are important for cell proliferation or apoptosis contain protein thiols, acting as centers for crosstalk between zinc- and redox-signaling. A prominent example for shared molecular targets for zinc and ROS are active site cysteine thiols in protein tyrosine phosphatases (PTP), their activity being downregulated by oxidation as well as zinc binding. Because zinc binding also protects PTP thiols form irreversible oxidation, there is a multi-faceted reciprocal interaction, illustrating that zinc- and redox-signaling are intricately linked on multiple levels.
Collapse
|
31
|
Hydrogen peroxide reactivity and specificity in thiol-based cell signalling. Biochem Soc Trans 2021; 48:745-754. [PMID: 32412042 DOI: 10.1042/bst20190049] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
Reversible oxidation of thiol proteins is an important cell signalling mechanism. In many cases, this involves generation or exposure of the cells to H2O2, and oxidation of proteins that are not particularly H2O2-reactive. There is a conundrum as to how these proteins are oxidised when other highly reactive proteins such as peroxiredoxins are present. This article discusses potential mechanisms, focussing on recent evidence for oxidation being localised within the cell, redox relays involving peroxiredoxins operating in some signalling pathways, and mechanisms for facilitated or directed oxidation of specific targets. These findings help define conditions that enable redox signalling but there is still much to learn regarding mechanisms.
Collapse
|
32
|
Wang M, Murdoch CE, Brewer AC, Ivetic A, Evans P, Shah AM, Zhang M. Endothelial NADPH oxidase 4 protects against angiotensin II-induced cardiac fibrosis and inflammation. ESC Heart Fail 2021; 8:1427-1437. [PMID: 33511759 PMCID: PMC8006688 DOI: 10.1002/ehf2.13228] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/07/2020] [Accepted: 01/13/2021] [Indexed: 12/21/2022] Open
Abstract
Aims Endothelial activation and inflammatory cell infiltration have important roles in the development of cardiac fibrosis induced by renin–angiotensin system activation. NADPH oxidases (Nox proteins) are expressed in endothelial cells (ECs) and alter their function. Previous studies indicated that Nox2 in ECs contributes to angiotensin II (AngII)‐induced cardiac fibrosis. However, the effects of EC Nox4 on cardiac fibrosis are unknown. Methods and results Transgenic (TG) mice overexpressing endothelial‐restricted Nox4 were studied alongside wild‐type (WT) littermates as controls. At baseline, Nox4 TG mice had significantly enlarged hearts compared with WT, with elongated cardiomyocytes (increased by 18.5%, P < 0.01) and eccentric hypertrophy but well‐preserved cardiac function by echocardiography and in vivo pressure–volume analysis. Animals were subjected to a chronic AngII infusion (AngII, 1.1 mg/kg/day) for 14 days. Whereas WT/AngII developed a 2.1‐fold increase in interstitial cardiac fibrosis as compared with WT/saline controls (P < 0.01), TG/AngII mice developed significant less fibrosis (1.4‐fold increase, P > 0.05), but there were no differences in cardiac hypertrophy or contractile function between the two groups. TG hearts displayed significantly decreased inflammatory cell infiltration with reduced levels of vascular cell adhesion molecule 1 in both the vasculature and myocardium compared with WT after AngII treatment. TG microvascular ECs stimulated with AngII in vitro supported significantly less leukocyte adhesion than WT ECs. Conclusions A chronic increase in endothelial Nox4 stimulates physiological cardiac hypertrophy and protects against AngII‐induced cardiac fibrosis by inhibiting EC activation and the recruitment of inflammatory cells. Mice with endothelium‐specific overexpression of Nox4 (EndoNox4 TG) exhibit eccentric hypertrophy with well‐preserved cardiac function at baseline. EndoNox4 TG mice develop significantly less interstitial cardiac fibrosis in response to chronic pressure AngII stimulation, independent of cardiac hypertrophy. Overexpression of Nox4 in endothelial cells reduces AngII‐induced endothelial activation. An increase in endothelial Nox4 inhibits AngII‐induced recruitment of inflammatory cells in the heart.
Collapse
Affiliation(s)
- Minshu Wang
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London, SE5 9NU, UK.,Department of Ophthalmology, Peking University Third Hospital, Beijing, China
| | - Colin E Murdoch
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Alison C Brewer
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Aleksandar Ivetic
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Paul Evans
- Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Sheffield, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Min Zhang
- School of Cardiovascular Medicine and Sciences, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London, SE5 9NU, UK
| |
Collapse
|
33
|
Konno T, Melo EP, Chambers JE, Avezov E. Intracellular Sources of ROS/H 2O 2 in Health and Neurodegeneration: Spotlight on Endoplasmic Reticulum. Cells 2021; 10:233. [PMID: 33504070 PMCID: PMC7912550 DOI: 10.3390/cells10020233] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/08/2023] Open
Abstract
Reactive oxygen species (ROS) are produced continuously throughout the cell as products of various redox reactions. Yet these products function as important signal messengers, acting through oxidation of specific target factors. Whilst excess ROS production has the potential to induce oxidative stress, physiological roles of ROS are supported by a spatiotemporal equilibrium between ROS producers and scavengers such as antioxidative enzymes. In the endoplasmic reticulum (ER), hydrogen peroxide (H2O2), a non-radical ROS, is produced through the process of oxidative folding. Utilisation and dysregulation of H2O2, in particular that generated in the ER, affects not only cellular homeostasis but also the longevity of organisms. ROS dysregulation has been implicated in various pathologies including dementia and other neurodegenerative diseases, sanctioning a field of research that strives to better understand cell-intrinsic ROS production. Here we review the organelle-specific ROS-generating and consuming pathways, providing evidence that the ER is a major contributing source of potentially pathologic ROS.
Collapse
Affiliation(s)
- Tasuku Konno
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Eduardo Pinho Melo
- CCMAR—Centro de Ciências do Mar, Campus de Gambelas, Universidade do Algarve, 8005-139 Faro, Portugal;
| | - Joseph E. Chambers
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK;
| | - Edward Avezov
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
34
|
Schnelle M, Sawyer I, Anilkumar N, Mohamed BA, Richards DA, Toischer K, Zhang M, Catibog N, Sawyer G, Mongue-Din H, Schröder K, Hasenfuss G, Shah AM. NADPH oxidase-4 promotes eccentric cardiac hypertrophy in response to volume overload. Cardiovasc Res 2021; 117:178-187. [PMID: 31821410 PMCID: PMC7797217 DOI: 10.1093/cvr/cvz331] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/13/2019] [Accepted: 12/07/2019] [Indexed: 12/15/2022] Open
Abstract
AIMS Chronic pressure or volume overload induce concentric vs. eccentric left ventricular (LV) remodelling, respectively. Previous studies suggest that distinct signalling pathways are involved in these responses. NADPH oxidase-4 (Nox4) is a reactive oxygen species-generating enzyme that can limit detrimental cardiac remodelling in response to pressure overload. This study aimed to assess its role in volume overload-induced remodelling. METHODS AND RESULTS We compared the responses to creation of an aortocaval fistula (Shunt) to induce volume overload in Nox4-null mice (Nox4-/-) vs. wild-type (WT) littermates. Induction of Shunt resulted in a significant increase in cardiac Nox4 mRNA and protein levels in WT mice as compared to Sham controls. Nox4-/- mice developed less eccentric LV remodelling than WT mice (echocardiographic relative wall thickness: 0.30 vs. 0.27, P < 0.05), with less LV hypertrophy at organ level (increase in LV weight/tibia length ratio of 25% vs. 43%, P < 0.01) and cellular level (cardiomyocyte cross-sectional area: 323 µm2 vs. 379 μm2, P < 0.01). LV ejection fraction, foetal gene expression, interstitial fibrosis, myocardial capillary density, and levels of myocyte apoptosis after Shunt were similar in the two genotypes. Myocardial phospho-Akt levels were increased after induction of Shunt in WT mice, whereas levels decreased in Nox4-/- mice (+29% vs. -21%, P < 0.05), associated with a higher level of phosphorylation of the S6 ribosomal protein (S6) and the eIF4E-binding protein 1 (4E-BP1) in WT compared to Nox4-/- mice. We identified that Akt activation in cardiac cells is augmented by Nox4 via a Src kinase-dependent inactivation of protein phosphatase 2A. CONCLUSION Endogenous Nox4 is required for the full development of eccentric cardiac hypertrophy and remodelling during chronic volume overload. Nox4-dependent activation of Akt and its downstream targets S6 and 4E-BP1 may be involved in this effect.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Apoptosis
- Arteriovenous Shunt, Surgical
- Cell Cycle Proteins/metabolism
- Cell Line
- Disease Models, Animal
- Fibrosis
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- NADPH Oxidase 2/genetics
- NADPH Oxidase 2/metabolism
- NADPH Oxidase 4/genetics
- NADPH Oxidase 4/metabolism
- Phosphorylation
- Protein Phosphatase 2/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Ribosomal Protein S6/metabolism
- Signal Transduction
- Ventricular Function, Left
- Ventricular Remodeling
- src-Family Kinases/metabolism
- Mice
Collapse
Affiliation(s)
- Moritz Schnelle
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- Institute for Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Iain Sawyer
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Narayana Anilkumar
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Daniel A Richards
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Min Zhang
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Norman Catibog
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Greta Sawyer
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Héloïse Mongue-Din
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Ajay M Shah
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
35
|
Beretta M, Santos CXC, Molenaar C, Hafstad AD, Miller CCJ, Revazian A, Betteridge K, Schröder K, Streckfuß‐Bömeke K, Doroshow JH, Fleck RA, Su T, Belousov VV, Parsons M, Shah AM. Nox4 regulates InsP 3 receptor-dependent Ca 2+ release into mitochondria to promote cell survival. EMBO J 2020; 39:e103530. [PMID: 33001475 PMCID: PMC7527947 DOI: 10.15252/embj.2019103530] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 06/22/2020] [Accepted: 07/01/2020] [Indexed: 12/24/2022] Open
Abstract
Cells subjected to environmental stresses undergo regulated cell death (RCD) when homeostatic programs fail to maintain viability. A major mechanism of RCD is the excessive calcium loading of mitochondria and consequent triggering of the mitochondrial permeability transition (mPT), which is especially important in post-mitotic cells such as cardiomyocytes and neurons. Here, we show that stress-induced upregulation of the ROS-generating protein Nox4 at the ER-mitochondria contact sites (MAMs) is a pro-survival mechanism that inhibits calcium transfer through InsP3 receptors (InsP3 R). Nox4 mediates redox signaling at the MAM of stressed cells to augment Akt-dependent phosphorylation of InsP3 R, thereby inhibiting calcium flux and mPT-dependent necrosis. In hearts subjected to ischemia-reperfusion, Nox4 limits infarct size through this mechanism. These results uncover a hitherto unrecognized stress pathway, whereby a ROS-generating protein mediates pro-survival effects through spatially confined signaling at the MAM to regulate ER to mitochondria calcium flux and triggering of the mPT.
Collapse
Affiliation(s)
- Matteo Beretta
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| | - Celio XC Santos
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| | - Chris Molenaar
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| | - Anne D Hafstad
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
- Cardiovascular Research GroupDepartment of Medical BiologyUIT‐The Arctic University of NorwayTromsøNorway
| | - Chris CJ Miller
- Department of Basic and Clinical NeuroscienceInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Aram Revazian
- Institute for Cardiovascular PhysiologyGeorg August University GöttingenGöttingenGermany
| | - Kai Betteridge
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| | - Katrin Schröder
- Institute for Cardiovascular PhysiologyGoethe‐University FrankfurtFrankfurt am MainGermany
| | | | - James H Doroshow
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteNIHBethesdaMDUSA
| | - Roland A Fleck
- Centre for Ultrastructural ImagingKing's College LondonLondonUK
| | - Tsung‐Ping Su
- Cellular Pathobiology SectionNational Institute on Drug Abuse, NIHBaltimoreMDUSA
| | - Vsevolod V Belousov
- Institute for Cardiovascular PhysiologyGeorg August University GöttingenGöttingenGermany
- Shemyakin‐Ovchinnikov Institute of Bioorganic ChemistryMoscowRussia
- Pirogov Russian National Research Medical UniversityMoscowRussia
| | - Maddy Parsons
- King's College London British Heart Foundation CentreRandall Centre of Cell and Molecular BiophysicsLondonUK
| | - Ajay M Shah
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| |
Collapse
|
36
|
Ireland SC, Huang H, Zhang J, Li J, Wang Y. Hydrogen peroxide induces Arl1 degradation and impairs Golgi-mediated trafficking. Mol Biol Cell 2020; 31:1931-1942. [PMID: 32583744 PMCID: PMC7525819 DOI: 10.1091/mbc.e20-01-0063] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/02/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS)-induced oxidative stress has been associated with diseases such as amyotrophic lateral sclerosis, stroke, and cancer. While the effect of ROS on mitochondria and endoplasmic reticulum (ER) has been well documented, its consequence on the Golgi apparatus is less well understood. In this study, we characterized the Golgi structure and function in HeLa cells after exposure to hydrogen peroxide (H2O2), a reagent commonly used to introduce ROS to cells. Treatment of cells with 1 mM H2O2 for 10 min resulted in the degradation of Arl1 and dissociation of GRIP domain-containing proteins Golgin-97 and Golgin-245 from the trans-Golgi. This effect could be rescued by treatment of cells with a ROS scavenger N-acetyl cysteine or protease inhibitors. Structurally, H2O2 treatment reduced the number of cisternal membranes per Golgi stack, suggesting a loss of trans-Golgi cisternae. Functionally, H2O2 treatment inhibited both anterograde and retrograde protein transport, consistent with the loss of membrane tethers on the trans-Golgi cisternae. This study revealed membrane tethers at the trans-Golgi as novel specific targets of ROS in cells.
Collapse
Affiliation(s)
- Stephen C. Ireland
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Haoran Huang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Jianchao Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109-1085
| |
Collapse
|
37
|
MnTE-2-PyP Suppresses Prostate Cancer Cell Growth via H 2O 2 Production. Antioxidants (Basel) 2020; 9:antiox9060490. [PMID: 32512786 PMCID: PMC7346125 DOI: 10.3390/antiox9060490] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/03/2020] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer patients are often treated with radiotherapy. MnTE-2-PyP, a superoxide dismutase (SOD) mimic, is a known radioprotector of normal tissues. Our recent work demonstrated that MnTE-2-PyP also inhibits prostate cancer progression with radiotherapy; however, the mechanisms remain unclear. In this study, we identified that MnTE-2-PyP-induced intracellular H2O2 levels are critical in inhibiting the growth of PC3 and LNCaP cells, but the increased H2O2 levels affected the two cancer cells differently. In PC3 cells, many proteins were thiol oxidized with MnTE-2-PyP treatment, including Ser/Thr protein phosphatase 1 beta catalytic subunit (PP1CB). This resulted in reduced PP1CB activity; however, overall cell cycle progression was not altered, so this is not the main mechanism of PC3 cell growth inhibition. High H2O2 levels by MnTE-2-PyP treatment induced nuclear fragmentation, which could be synergistically enhanced with radiotherapy. In LNCaP cells, thiol oxidation by MnTE-2-PyP treatment was not observed previously and, similarly to PC3 cells, there was no effect of MnTE-2-PyP treatment on cell cycle progression. However, in LNCaP cells, MnTE-2-PyP caused an increase in low RNA population and sub-G1 population of cells, which indicates that MnTE-2-PyP treatment may cause cellular quiescence or direct cancer cell death. The protein oxidative modifications and mitotic catastrophes caused by MnTE-2-PyP may be the major contributors to cell growth inhibition in PC3 cells, while in LNCaP cells, tumor cell quiescence or cell death appears to be major factors in MnTE-2-PyP-induced growth inhibition.
Collapse
|
38
|
Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol 2020; 21:363-383. [PMID: 32231263 DOI: 10.1038/s41580-020-0230-3] [Citation(s) in RCA: 2301] [Impact Index Per Article: 575.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
'Reactive oxygen species' (ROS) is an umbrella term for an array of derivatives of molecular oxygen that occur as a normal attribute of aerobic life. Elevated formation of the different ROS leads to molecular damage, denoted as 'oxidative distress'. Here we focus on ROS at physiological levels and their central role in redox signalling via different post-translational modifications, denoted as 'oxidative eustress'. Two species, hydrogen peroxide (H2O2) and the superoxide anion radical (O2·-), are key redox signalling agents generated under the control of growth factors and cytokines by more than 40 enzymes, prominently including NADPH oxidases and the mitochondrial electron transport chain. At the low physiological levels in the nanomolar range, H2O2 is the major agent signalling through specific protein targets, which engage in metabolic regulation and stress responses to support cellular adaptation to a changing environment and stress. In addition, several other reactive species are involved in redox signalling, for instance nitric oxide, hydrogen sulfide and oxidized lipids. Recent methodological advances permit the assessment of molecular interactions of specific ROS molecules with specific targets in redox signalling pathways. Accordingly, major advances have occurred in understanding the role of these oxidants in physiology and disease, including the nervous, cardiovascular and immune systems, skeletal muscle and metabolic regulation as well as ageing and cancer. In the past, unspecific elimination of ROS by use of low molecular mass antioxidant compounds was not successful in counteracting disease initiation and progression in clinical trials. However, controlling specific ROS-mediated signalling pathways by selective targeting offers a perspective for a future of more refined redox medicine. This includes enzymatic defence systems such as those controlled by the stress-response transcription factors NRF2 and nuclear factor-κB, the role of trace elements such as selenium, the use of redox drugs and the modulation of environmental factors collectively known as the exposome (for example, nutrition, lifestyle and irradiation).
Collapse
Affiliation(s)
- Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany. .,Leibniz Research Institute for Environmental Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Dean P Jones
- Department of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
39
|
Kalish JM, Tang XH, Scognamiglio T, Zhang T, Gudas LJ. Doxycycline-induced exogenous Bmi-1 expression enhances tumor formation in a murine model of oral squamous cell carcinoma. Cancer Biol Ther 2020; 21:400-411. [PMID: 32037955 DOI: 10.1080/15384047.2020.1720485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
B Cell-Specific Moloney Murine Leukemia Virus Integration Site 1 (Bmi-1, Bmi1), an epigenetic protein, is necessary for normal stem cell self-renewal in adult animals and for cancer stem cell (CSC) functions in adult animals. To elucidate the functions of Bmi-1 in the oral cavity we created a transgenic mouse line (KrTBmi-1) that expresses ectopic, Flag-tagged Bmi-1 in tongue basal epithelial stem cells only upon doxycycline (DOX) treatment. Genome wide transcriptomics and Ingenuity Pathway Analysis identified several pathways altered by exogenous Bmi-1 expression in the normal tongue epithelium, including EIF2 signaling (P value = 1.58 x 10-49), mTOR signaling (P value = 2.45 x 10-12), oxidative phosphorylation (P = 6.61 x 10-3) and glutathione redox reactions I (P = 1.74 x 10-2). Overall, our data indicate that ectopic Bmi-1 expression has an impact on normal tongue epithelial homeostasis. We then assessed the KrTBmi-1 mice in the 4-nitroquinoline 1-oxide (4-NQO) model of oral carcinogenesis. We found that 80% of mice expressing exogenous Bmi-1 (+DOX, +4-NQO KrTBmi-1; N = 10) developed tumors classified as grade 3 or higher, compared to 60% and 40% of mice expressing just endogenous Bmi-1 (+DOX, +4-NQO Kr and -DOX, +4-NQO KrTBmi-1 groups, respectively; N = 10/group; P value = <0.0001); and 30% of mice expressing ectopic Bmi-1 mice developed 20 or more lesions compared to 10% of mice expressing only endogenous Bmi-1 (P = .009). This demonstrates that exogenous Bmi-1 expression increases the susceptibility of mice to 4-NQO-induced oral carcinogenesis, strengthening the evidence for Bmi-1 as a therapeutic target in human oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Jocelin M Kalish
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA.,Department of Pharmacology, Weill Cornell Graduate School of Biomedical Sciences, New York, NY, USA.,Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA.,Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | | | - Tuo Zhang
- Weill Cornell Genomics Core Facility, Weill Cornell Medical College, New York, NY, USA
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA.,Department of Pharmacology, Weill Cornell Graduate School of Biomedical Sciences, New York, NY, USA.,Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
40
|
Rajaram RD, Dissard R, Jaquet V, de Seigneux S. Potential benefits and harms of NADPH oxidase type 4 in the kidneys and cardiovascular system. Nephrol Dial Transplant 2020; 34:567-576. [PMID: 29931336 DOI: 10.1093/ndt/gfy161] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Indexed: 12/21/2022] Open
Abstract
The main function of NADPH oxidases is to catalyse the formation of reactive oxygen species (ROS). NADPH oxidase 4 (NOX4) is expressed at high levels in kidney tubular cells, and at lower levels in endothelial cells, cardiomyocytes and other cell types under physiological conditions. NOX4 is constitutively active producing hydrogen peroxide (H2O2) as the prevalent ROS detected, whereas other NOX isoforms present in the renal and cardiovascular systems (i.e. NOX1, NOX2 and NOX5) generate superoxide radical anions as main products. Pharmacological inhibition of NOX4 has received enormous attention for its potential therapeutic benefit in fibrotic disease and nephropathologies. Ongoing clinical trials are testing this approach in humans. Diabetes elevates NOX4 expression in podocytes and mesangial cells, which was shown to damage glomeruli leading to podocyte loss, mesangial cell hypertrophy and matrix accumulation. Consequently, NOX4 represents an interesting therapeutic target in diabetic nephropathy. On the contrary, experiments using NOX4-deficient mice have shown that NOX4 is cytoprotective in tubular cells, cardiomyocytes, endothelial cells and vascular smooth muscle cells, and has a metabolism-regulating role when these cells are subjected to injury. Mice with systemic NOX4 deletion are more susceptible to acute and chronic tubular injury, heart failure and atherosclerosis. Overall, the current literature suggests a detrimental role of increased NOX4 expression in mesangial cells and podocytes during diabetic nephropathy, but a cytoprotective role of this enzyme in other cellular types where it is expressed endogenously. We review here the recent evidence on the role of NOX4 in the kidneys and cardiovascular system. With the emergence of pharmacological NOX4 inhibitors in clinical trials, caution should be taken in identifying potential side effects in patients prone to acute kidney injury and cardiovascular disease.
Collapse
Affiliation(s)
- Renuga D Rajaram
- Laboratory of Nephrology, Service of Nephrology, Departments of Internal Medicine Specialties and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Romain Dissard
- Laboratory of Nephrology, Service of Nephrology, Departments of Internal Medicine Specialties and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Vincent Jaquet
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sophie de Seigneux
- Laboratory of Nephrology, Service of Nephrology, Departments of Internal Medicine Specialties and Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
41
|
Delitsikou V, Jarad G, Rajaram RD, Ino F, Rutkowski JM, Chen CD, Santos CXC, Scherer PE, Abraham CR, Shah AM, Feraille E, Miner JH, de Seigneux S. Klotho regulation by albuminuria is dependent on ATF3 and endoplasmic reticulum stress. FASEB J 2019; 34:2087-2104. [PMID: 31907991 DOI: 10.1096/fj.201900893r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 10/31/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
Abstract
Proteinuria is associated with renal function decline and cardiovascular mortality. This association may be attributed in part to alterations of Klotho expression induced by albuminuria, yet the underlying mechanisms are unclear. The presence of albumin decreased Klotho expression in the POD-ATTAC mouse model of proteinuric kidney disease as well as in kidney epithelial cell lines. This downregulation was related to both decreased Klotho transcription and diminished protein half-life, whereas cleavage by ADAM proteases was not modified. The regulation was albumin specific since it was neither observed in the analbuminemic Col4α3-/- Alport mice nor induced by exposure of kidney epithelial cells to purified immunoglobulins. Albumin induced features of ER stress in renal tubular cells with ATF3/ATF4 activation. ATF3 and ATF4 induction downregulated Klotho through altered transcription mediated by their binding on the Klotho promoter. Inhibiting ER stress with 4-PBA decreased the effect of albumin on Klotho protein levels without altering mRNA levels, thus mainly abrogating the increased protein degradation. Taken together, albuminuria decreases Klotho expression through increased protein degradation and decreased transcription mediated by ER stress induction. This implies that modulating ER stress may improve proteinuria-induced alterations of Klotho expression, and hence renal and extrarenal complications associated with Klotho loss.
Collapse
Affiliation(s)
- Vasiliki Delitsikou
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland.,Laboratory of Nephrology, Department of Internal Medicine Specialties, HUG, Geneva, Switzerland
| | - George Jarad
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Renuga Devi Rajaram
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland.,Laboratory of Nephrology, Department of Internal Medicine Specialties, HUG, Geneva, Switzerland
| | - Frédérique Ino
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland.,Laboratory of Nephrology, Department of Internal Medicine Specialties, HUG, Geneva, Switzerland
| | - Joseph M Rutkowski
- Touchstone Diabetes Centre, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Medical Physiology, Texas A&M College of Medicine, College Station, Texas
| | - Ci-Di Chen
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Celio X C Santos
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Philipp E Scherer
- Touchstone Diabetes Centre, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Carmela R Abraham
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Eric Feraille
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland
| | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Sophie de Seigneux
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland.,Laboratory of Nephrology, Department of Internal Medicine Specialties, HUG, Geneva, Switzerland
| |
Collapse
|
42
|
Tsushima M, Liu J, Hirao W, Yamazaki H, Tomita H, Itoh K. Emerging evidence for crosstalk between Nrf2 and mitochondria in physiological homeostasis and in heart disease. Arch Pharm Res 2019; 43:286-296. [DOI: 10.1007/s12272-019-01188-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/01/2019] [Indexed: 12/31/2022]
|
43
|
Zhang X, Gibhardt CS, Will T, Stanisz H, Körbel C, Mitkovski M, Stejerean I, Cappello S, Pacheu‐Grau D, Dudek J, Tahbaz N, Mina L, Simmen T, Laschke MW, Menger MD, Schön MP, Helms V, Niemeyer BA, Rehling P, Vultur A, Bogeski I. Redox signals at the ER-mitochondria interface control melanoma progression. EMBO J 2019; 38:e100871. [PMID: 31304984 PMCID: PMC6669928 DOI: 10.15252/embj.2018100871] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen species (ROS) are emerging as important regulators of cancer growth and metastatic spread. However, how cells integrate redox signals to affect cancer progression is not fully understood. Mitochondria are cellular redox hubs, which are highly regulated by interactions with neighboring organelles. Here, we investigated how ROS at the endoplasmic reticulum (ER)-mitochondria interface are generated and translated to affect melanoma outcome. We show that TMX1 and TMX3 oxidoreductases, which promote ER-mitochondria communication, are upregulated in melanoma cells and patient samples. TMX knockdown altered mitochondrial organization, enhanced bioenergetics, and elevated mitochondrial- and NOX4-derived ROS. The TMX-knockdown-induced oxidative stress suppressed melanoma proliferation, migration, and xenograft tumor growth by inhibiting NFAT1. Furthermore, we identified NFAT1-positive and NFAT1-negative melanoma subgroups, wherein NFAT1 expression correlates with melanoma stage and metastatic potential. Integrative bioinformatics revealed that genes coding for mitochondrial- and redox-related proteins are under NFAT1 control and indicated that TMX1, TMX3, and NFAT1 are associated with poor disease outcome. Our study unravels a novel redox-controlled ER-mitochondria-NFAT1 signaling loop that regulates melanoma pathobiology and provides biomarkers indicative of aggressive disease.
Collapse
Affiliation(s)
- Xin Zhang
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
- BiophysicsCIPMMSaarland UniversityHomburgGermany
| | - Christine S Gibhardt
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Thorsten Will
- Center for BioinformaticsSaarland UniversitySaarbrückenGermany
| | - Hedwig Stanisz
- Department of Dermatology, Venereology and AllergologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Christina Körbel
- Institute for Clinical and Experimental SurgerySaarland UniversityHomburgGermany
| | - Miso Mitkovski
- Light Microscopy FacilityMax Planck Institute for Experimental MedicineGöttingenGermany
| | - Ioana Stejerean
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Sabrina Cappello
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - David Pacheu‐Grau
- Department of Cellular BiochemistryUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Jan Dudek
- Department of Cellular BiochemistryUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Nasser Tahbaz
- Department of Cell BiologyUniversity of AlbertaEdmontonABCanada
| | - Lucas Mina
- Department of Cell BiologyUniversity of AlbertaEdmontonABCanada
| | - Thomas Simmen
- Department of Cell BiologyUniversity of AlbertaEdmontonABCanada
| | - Matthias W Laschke
- Institute for Clinical and Experimental SurgerySaarland UniversityHomburgGermany
| | - Michael D Menger
- Institute for Clinical and Experimental SurgerySaarland UniversityHomburgGermany
| | - Michael P Schön
- Department of Dermatology, Venereology and AllergologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Volkhard Helms
- Center for BioinformaticsSaarland UniversitySaarbrückenGermany
| | | | - Peter Rehling
- Department of Cellular BiochemistryUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
- Max Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Adina Vultur
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Ivan Bogeski
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
- BiophysicsCIPMMSaarland UniversityHomburgGermany
| |
Collapse
|
44
|
Gray SP, Shah AM, Smyrnias I. NADPH oxidase 4 and its role in the cardiovascular system. ACTA ACUST UNITED AC 2019; 1:H59-H66. [PMID: 32923955 PMCID: PMC7439918 DOI: 10.1530/vb-19-0014] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/11/2019] [Indexed: 12/19/2022]
Abstract
The heart relies on complex mechanisms that provide adequate myocardial oxygen supply in order to maintain its contractile function. At the cellular level, oxygen undergoes one electron reduction to superoxide through the action of different types of oxidases (e.g. xanthine oxidases, uncoupled nitric oxide synthases, NADPH oxidases or NOX). Locally generated oxygen-derived reactive species (ROS) are involved in various signaling pathways including cardiac adaptation to different types of physiological and pathophysiological stresses (e.g. hypoxia or overload). The specific effects of ROS and their regulation by oxidases are dependent on the amount of ROS generated and their specific subcellular localization. The NOX family of NADPH oxidases is a main source of ROS in the heart. Seven distinct Nox isoforms (NOX1–NOX5 and DUOX1 and 2) have been identified, of which NOX1, 2, 4 and 5 have been characterized in the cardiovascular system. For the purposes of this review, we will focus on the effects of NADPH oxidase 4 (NOX4) in the heart.
Collapse
Affiliation(s)
- Stephen P Gray
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre, London, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre, London, UK
| | - Ioannis Smyrnias
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre, London, UK
| |
Collapse
|
45
|
Zhang M, Mongue-Din H, Martin D, Catibog N, Smyrnias I, Zhang X, Yu B, Wang M, Brandes RP, Schröder K, Shah AM. Both cardiomyocyte and endothelial cell Nox4 mediate protection against hemodynamic overload-induced remodelling. Cardiovasc Res 2019; 114:401-408. [PMID: 29040462 PMCID: PMC6018755 DOI: 10.1093/cvr/cvx204] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 10/12/2017] [Indexed: 12/27/2022] Open
Abstract
Aims NADPH oxidase-4 (Nox4) is an important reactive oxygen species (ROS) source that is upregulated in the haemodynamically overloaded heart. Our previous studies using global Nox4 knockout (Nox4KO) mice demonstrated a protective role of Nox4 during chronic abdominal aortic banding, involving a paracrine enhancement of myocardial capillary density. However, other authors who studied cardiac-specific Nox4KO mice reported detrimental effects of Nox4 in response to transverse aortic constriction (TAC). It has been speculated that these divergent results are due to cell-specific actions of Nox4 (i.e. cardiomyocyte Nox4 detrimental but endothelial Nox4 beneficial) and/or differences in the model of pressure overload (i.e. abdominal banding vs. TAC). This study aimed to (i) investigate whether the effects of Nox4 on pressure overload-induced cardiac remodelling vary according to the pressure overload model and (ii) compare the roles of cardiomyocyte vs. endothelial cell Nox4. Methods and results Global Nox4KO mice subjected to TAC developed worse cardiac remodelling and contractile dysfunction than wild-type littermates, consistent with our previous results with abdominal aortic banding. Next, we generated inducible cardiomyocyte-specific Nox4 KO mice (Cardio-Nox4KO) and endothelial-specific Nox4 KO mice (Endo-Nox4KO) and studied their responses to pressure overload. Both Cardio-Nox4KO and Endo-Nox4KO developed worse pressure overload-induced cardiac remodelling and dysfunction than wild-type littermates, associated with significant decrease in protein levels of HIF1α and VEGF and impairment of myocardial capillarization. Conclusions Cardiomyocyte as well as endothelial cell Nox4 contributes to protection against chronic hemodynamic overload-induced cardiac remodelling, at least in part through common effects on myocardial capillary density.
Collapse
Affiliation(s)
- Min Zhang
- Cardiovascular Division, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Heloise Mongue-Din
- Cardiovascular Division, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Daniel Martin
- Cardiovascular Division, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Norman Catibog
- Cardiovascular Division, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Ioannis Smyrnias
- Cardiovascular Division, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Xiaohong Zhang
- Cardiovascular Division, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Bin Yu
- Cardiovascular Division, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Minshu Wang
- Cardiovascular Division, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Ajay M Shah
- Cardiovascular Division, James Black Centre, King's College London British Heart Foundation Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
46
|
Inhibition of protein phosphatase 1 stimulates noncanonical ER stress eIF2α activation to enhance fisetin-induced chemosensitivity in HDAC inhibitor-resistant hepatocellular carcinoma cells. Cancers (Basel) 2019; 11:cancers11070918. [PMID: 31261976 PMCID: PMC6678694 DOI: 10.3390/cancers11070918] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/14/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common fatal type of malignant tumor that has highly metastatic and recurrent properties. Fisetin is a natural flavonoid found in various vegetables and fruits which exhibits anti-cancer and anti-inflammatory properties, as well as other effects. Thus, we hypothesized that fisetin can act as an adjuvant therapy in cancer or drug-resistant cancer cells, and further investigated the molecular mechanisms underlying the development of drug-resistance in HCC cells. We found that fisetin effectively inhibited the cell viability of not only parental cells but also histone deacetylase inhibitors-resistant (HDACis-R) cells and enhanced the chemosensitivity of HCC cells. Interestingly, fisetin did not induce cell apoptosis through the activation of the endoplasmic reticulum (ER) stress sensor of protein kinase R (PKR)-like endoplasmic reticulum kinase, but rather through the non-canonical pathway of the protein phosphatase 1 (PP1)-mediated suppression of eIF2α phosphorylation. Moreover, fisetin-induced cell apoptosis was reversed by treatment with PP1 activator or eIF2α siRNA in HCC cells. Based on these observations, we suggest that PP1-eIF2α pathways are significantly involved in the effect of fisetin on HCC apoptosis. Thus, fisetin may act as a novel anticancer drug and new chemotherapy adjuvant which can improve the efficacy of chemotherapeutic agents and diminish their side-effects.
Collapse
|
47
|
Rajaram RD, Dissard R, Faivre A, Ino F, Delitsikou V, Jaquet V, Cagarelli T, Lindenmeyer M, Jansen-Duerr P, Cohen C, Moll S, de Seigneux S. Tubular NOX4 expression decreases in chronic kidney disease but does not modify fibrosis evolution. Redox Biol 2019; 26:101234. [PMID: 31247506 PMCID: PMC6598841 DOI: 10.1016/j.redox.2019.101234] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/27/2019] [Accepted: 05/31/2019] [Indexed: 12/19/2022] Open
Abstract
Background NADPH oxidase 4 (NOX4) catalyzes the formation of hydrogen peroxide (H2O2). NOX4 is highly expressed in the kidney, but its role in renal injury is unclear and may depend on its specific tissue localization. Methods We performed immunostaining with a specific anti-NOX4 antibody and measured NOX4 mRNA expression in human renal biopsies encompassing diverse renal diseases. We generated transgenic mice specifically overexpressing mouse Nox4 in renal tubular cells and subjected the animals to the unilateral ureteral obstruction (UUO) model of fibrosis. Results In normal human kidney, NOX4 protein expression was at its highest on the basolateral side of proximal tubular cells. NOX4 expression increased in mesangial cells and podocytes in proliferative diabetic nephropathy. In tubular cells, NOX4 protein expression decreased in all types of chronic renal disease studied. This finding was substantiated by decreased NOX4 mRNA expression in the tubulo-interstitial compartment in a repository of 175 human renal biopsies. Overexpression of tubular NOX4 in mice resulted in enhanced renal production of H2O2, increased NRF2 protein expression and decreased glomerular filtration, likely via stimulation of the tubulo-glomerular feedback. Tubular NOX4 overexpression had no obvious impact on kidney morphology, apoptosis, or fibrosis at baseline. Under acute and chronic tubular injury induced by UUO, overexpression of NOX4 in tubular cells did not modify the course of the disease. Conclusions NOX4 expression was decreased in tubular cells in all types of CKD tested. Tubular NOX4 overexpression did not induce injury in the kidney, and neither modified microvascularization, nor kidney structural lesions in fibrosis.
Collapse
Affiliation(s)
- Renuga Devi Rajaram
- Laboratory of Nephrology, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland; Service of Nephrology, Department of Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Romain Dissard
- Laboratory of Nephrology, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland; Service of Nephrology, Department of Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Anna Faivre
- Laboratory of Nephrology, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland; Service of Nephrology, Department of Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Frédérique Ino
- Laboratory of Nephrology, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland; Service of Nephrology, Department of Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Vasiliki Delitsikou
- Laboratory of Nephrology, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland; Service of Nephrology, Department of Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
| | - Vincent Jaquet
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Thomas Cagarelli
- Service of Clinical Pathology, Department of Pathology and Immunology, University Hospital and University of Geneva, Geneva, Switzerland
| | - Maja Lindenmeyer
- Nephrological Center Medical Clinic and Polyclinic IV, University of Munich, Munich, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pidder Jansen-Duerr
- Universität Innsbruck, Research Institute for Biomedical Aging Research, Rennweg 10, Innsbruck, Austria
| | - Clemens Cohen
- Nephrological Center Medical Clinic and Polyclinic IV, University of Munich, Munich, Germany
| | - Solange Moll
- Service of Clinical Pathology, Department of Pathology and Immunology, University Hospital and University of Geneva, Geneva, Switzerland
| | - Sophie de Seigneux
- Laboratory of Nephrology, Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland; Service of Nephrology, Department of Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland.
| |
Collapse
|
48
|
Paracrine Mechanisms of Redox Signalling for Postmitotic Cell and Tissue Regeneration. Trends Cell Biol 2019; 29:514-530. [DOI: 10.1016/j.tcb.2019.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 01/08/2023]
|
49
|
H 2O 2 Metabolism in Normal Thyroid Cells and in Thyroid Tumorigenesis: Focus on NADPH Oxidases. Antioxidants (Basel) 2019; 8:antiox8050126. [PMID: 31083324 PMCID: PMC6563055 DOI: 10.3390/antiox8050126] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/18/2019] [Accepted: 04/24/2019] [Indexed: 12/23/2022] Open
Abstract
Thyroid hormone synthesis requires adequate hydrogen peroxide (H2O2) production that is utilized as an oxidative agent during the synthesis of thyroxin (T4) and triiodothyronine (T3). Thyroid H2O2 is generated by a member of the family of NADPH oxidase enzymes (NOX-es), termed dual oxidase 2 (DUOX2). NOX/DUOX enzymes produce reactive oxygen species (ROS) as their unique enzymatic activity in a timely and spatially regulated manner and therefore, are important regulators of diverse physiological processes. By contrast, dysfunctional NOX/DUOX-derived ROS production is associated with pathological conditions. Inappropriate DUOX2-generated H2O2 production results in thyroid hypofunction in rodent models. Recent studies also indicate that ROS improperly released by NOX4, another member of the NOX family, are involved in thyroid carcinogenesis. This review focuses on the current knowledge concerning the redox regulation of thyroid hormonogenesis and cancer development with a specific emphasis on the NOX and DUOX enzymes in these processes.
Collapse
|
50
|
Elgenaidi IS, Spiers JP. Hypoxia modulates protein phosphatase 2A through HIF-1α dependent and independent mechanisms in human aortic smooth muscle cells and ventricular cardiomyocytes. Br J Pharmacol 2019; 176:1745-1763. [PMID: 30825189 DOI: 10.1111/bph.14648] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 02/05/2019] [Accepted: 02/13/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Although protein phosphatases regulate multiple cellular functions, their modulation under hypoxia remains unclear. We investigated expression of the protein phosphatase system under normoxic/hypoxic conditions and the mechanism by which hypoxia alters protein phosphatase 2A (PP2A) activity. EXPERIMENTAL APPROACH Human cardiovascular cells were cultured in cell type specific media under normoxic or hypoxic conditions (1% O2 ). Effects on mRNA expression, phosphatase activity, post-translational modification, and involvement of hypoxia inducible factor 1α (HIF-1α) were assessed using RT-PCR, immunoblotting, an activity assay, and siRNA silencing. KEY RESULTS All components of the protein phosphatase system studied were expressed in each cell line. Hypoxia attenuated mRNA expression of the transcripts in a cell line- and time-dependent manner. In human aortic smooth muscle cells (HASMC) and AC16 cells, hypoxia decreased PP2Ac activity and mRNA expression without altering PP2Ac abundance. Hypoxia increased demethylated PP2Ac (DPP2Ac) and phosphatase methylesterase 1 (PME-1) abundance but decreased leucine carboxyl methyltransferase 1 (LCMT-1) abundance. HIF-1α siRNA prevented the hypoxia-mediated decrease in phosphatase activity and expression of the catalytic subunit of protein phosphatase 2A (PPP2CA), independently of altering pPP2Ac, DPP2Ac, LCMT-1, or PME-1 abundance. CONCLUSION AND IMPLICATIONS Cardiovascular cells express multiple components of the PP2A system. In HASMC and AC16 cells, hypoxia inhibits PP2A activity through HIF-1α-dependent and -independent mechanisms, with the latter being consistent with altered PP2A holoenzyme assembly. This indicates a complex inhibitory effect of hypoxia on the PP2A system, and highlights PP2A as a therapeutic target for diseases associated with dysregulated protein phosphorylation.
Collapse
Affiliation(s)
| | - James Paul Spiers
- Department of Pharmacology and Therapeutics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|