1
|
Zhang H, Hao J, Hong H, Gu W, Li Z, Sun J, Zhan H, Wei X, Zhou L. Redox signaling regulates the skeletal tissue development and regeneration. Biotechnol Genet Eng Rev 2024; 40:2308-2331. [PMID: 37043672 DOI: 10.1080/02648725.2023.2199244] [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: 03/09/2023] [Accepted: 03/30/2023] [Indexed: 04/14/2023]
Abstract
Skeletal tissue development and regeneration in mammals are intricate, multistep, and highly regulated processes. Various signaling pathways have been implicated in the regulation of these processes, including redox. Redox signaling is the signal transduction by electron transfer reactions involving free radicals or related species. Redox homeostasis is essential to cell metabolic states, as the ROS not only regulates cell biological processes but also mediates physiological processes. Following a bone fracture, redox signaling is also triggered to regulate bone healing and regeneration by targeting resident stromal cells, osteoblasts, osteoclasts and endothelial cells. This review will focus on how the redox signaling impact the bone development and bone regeneration.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - Jin Hao
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - HaiPing Hong
- FangTa Hospital of Traditional Chinese Medicine, Songjiang Branch, Shanghai, East China, China
| | - Wei Gu
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | | | - Jun Sun
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - Hongsheng Zhan
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - Xiaoen Wei
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| | - Lin Zhou
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, East China, Shanghai, China
| |
Collapse
|
2
|
Hebchen DM, Schröder K. Redox Signaling in Endosomes Using the Example of EGF Receptors: A Graphical Review. Antioxidants (Basel) 2024; 13:1215. [PMID: 39456468 PMCID: PMC11504029 DOI: 10.3390/antiox13101215] [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: 09/22/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Early endosomes represent first-line sorting compartments or even organelles for internalized molecules. They enable the transport of molecules or ligands to other compartments of the cell, such as lysosomes, for degradation or recycle them back to the membrane by various mechanisms. Moreover, early endosomes function as signaling and scaffolding platforms to initiate or prolong distinct signaling pathways. Accordingly, early endosomes have to be recognized as either part of a degradation or recycling pathway. The physical proximity of many ligand-binding receptors with other membrane-bound proteins or complexes such as NADPH oxidases may result in an interaction of second messengers, like reactive oxygen species (ROS) and early endosomes, that promote the correct recognition of individual early endosomes. In fact, redoxosomes comprise an endosomal subsection of signaling endosomes. One example of such potential interaction is epidermal growth factor receptor (EGFR) signaling. Here we summarize recent findings on EGFR signaling as a well-studied example for receptor trafficking and trans-activation and illustrate the interplay between cellular and endosomal ROS.
Collapse
Affiliation(s)
| | - Katrin Schröder
- Institute of Physiology, Medical Faculty, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| |
Collapse
|
3
|
Schröder K. Specific signaling by nicotinamide adenine dinucleotide oxidases - Role of their site of action. Curr Opin Chem Biol 2024; 81:102461. [PMID: 38810503 DOI: 10.1016/j.cbpa.2024.102461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 05/31/2024]
Abstract
Nicotinamide adenine dinucleotide (NADPH) oxidases, known for their role in generating reactive oxygen species (ROS) have emerged as key regulators of specific cellular signaling pathways. While their primary function is ROS production, recent research has highlighted the significance of their site-specific activity in governing distinct cellular signaling events. NADPH oxidases (Nox) are found in various cell types, and both their expression and activities are tightly regulated. The generated ROS, such as superoxide anions and hydrogen peroxide, function as secondary messengers that modulate various signaling molecules, including protein kinases, transcription factors, and phosphatases. The site-specific action of NADPH oxidases in different cellular compartments, such as the plasma membrane, endosomes, and endoplasmic reticulum, allows for precise control over specific signaling pathways. Understanding the complex interplay of NADPH oxidases in cellular signaling is essential for deciphering their roles in health and disease. Dysregulation of these enzymes can lead to oxidative stress and inflammation, making them potential therapeutic targets in various pathological conditions. Ongoing research into NADPH oxidase activation and site-specific signaling promises to unveil new insights into cellular physiology and potential treatment strategies.
Collapse
|
4
|
Ma S, Xie X, Yuan R, Xin Q, Miao Y, Leng SX, Chen K, Cong W. Vascular Aging and Atherosclerosis: A Perspective on Aging. Aging Dis 2024; 16:AD.2024.0201-1. [PMID: 38502584 PMCID: PMC11745439 DOI: 10.14336/ad.2024.0201-1] [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: 10/25/2023] [Accepted: 02/01/2024] [Indexed: 03/21/2024] Open
Abstract
Vascular aging (VA) is recognized as a pivotal factor in the development and progression of atherosclerosis (AS). Although various epidemiological and clinical research has demonstrated an intimate connection between aging and AS, the candidate mechanisms still require thorough examination. This review adopts an aging-centric perspective to deepen the comprehension of the intricate relationship between biological aging, vascular cell senescence, and AS. Various aging-related physiological factors influence the physical system's reactions, including oxygen radicals, inflammation, lipids, angiotensin II, mechanical forces, glucose levels, and insulin resistance. These factors cause endothelial dysfunction, barrier damage, sclerosis, and inflammation for VA and promote AS via distinct or shared pathways. Furthermore, the increase of senescent cells inside the vascular tissues, caused by genetic damage, dysregulation, secretome changes, and epigenetic modifications, might be the primary cause of VA.
Collapse
Affiliation(s)
- Shudong Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xuena Xie
- School of Pharmacy, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Sean Xiao Leng
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA.
| | - Keji Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Weihong Cong
- School of Pharmacy, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
5
|
Ye W, Wang J, Little PJ, Zou J, Zheng Z, Lu J, Yin Y, Liu H, Zhang D, Liu P, Xu S, Ye W, Liu Z. Anti-atherosclerotic effects and molecular targets of ginkgolide B from Ginkgo biloba. Acta Pharm Sin B 2024; 14:1-19. [PMID: 38239238 PMCID: PMC10792990 DOI: 10.1016/j.apsb.2023.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/03/2023] [Accepted: 09/13/2023] [Indexed: 01/22/2024] Open
Abstract
Bioactive compounds derived from herbal medicinal plants modulate various therapeutic targets and signaling pathways associated with cardiovascular diseases (CVDs), the world's primary cause of death. Ginkgo biloba , a well-known traditional Chinese medicine with notable cardiovascular actions, has been used as a cardio- and cerebrovascular therapeutic drug and nutraceutical in Asian countries for centuries. Preclinical studies have shown that ginkgolide B, a bioactive component in Ginkgo biloba , can ameliorate atherosclerosis in cultured vascular cells and disease models. Of clinical relevance, several clinical trials are ongoing or being completed to examine the efficacy and safety of ginkgolide B-related drug preparations in the prevention of cerebrovascular diseases, such as ischemia stroke. Here, we present a comprehensive review of the pharmacological activities, pharmacokinetic characteristics, and mechanisms of action of ginkgolide B in atherosclerosis prevention and therapy. We highlight new molecular targets of ginkgolide B, including nicotinamide adenine dinucleotide phosphate oxidases (NADPH oxidase), lectin-like oxidized LDL receptor-1 (LOX-1), sirtuin 1 (SIRT1), platelet-activating factor (PAF), proprotein convertase subtilisin/kexin type 9 (PCSK9) and others. Finally, we provide an overview and discussion of the therapeutic potential of ginkgolide B and highlight the future perspective of developing ginkgolide B as an effective therapeutic agent for treating atherosclerosis.
Collapse
Affiliation(s)
- Weile Ye
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Jiaojiao Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Peter J. Little
- Pharmacy Australia Centre of Excellence, School of Pharmacy, University of Queensland, Woolloongabba QLD 4102, Australia
- Sunshine Coast Health Institute and School of Health and Behavioural Sciences, University of the Sunshine Coast, Birtinya QLD 4575, Australia
| | - Jiami Zou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Zhihua Zheng
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Jing Lu
- National-Local Joint Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Yanjun Yin
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Hao Liu
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Peiqing Liu
- National-Local Joint Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Suowen Xu
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, China
- Institute of Endocrine and Metabolic Diseases, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Wencai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Zhiping Liu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| |
Collapse
|
6
|
The mechanism and therapy of aortic aneurysms. Signal Transduct Target Ther 2023; 8:55. [PMID: 36737432 PMCID: PMC9898314 DOI: 10.1038/s41392-023-01325-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/15/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Aortic aneurysm is a chronic aortic disease affected by many factors. Although it is generally asymptomatic, it poses a significant threat to human life due to a high risk of rupture. Because of its strong concealment, it is difficult to diagnose the disease in the early stage. At present, there are no effective drugs for the treatment of aneurysms. Surgical intervention and endovascular treatment are the only therapies. Although current studies have discovered that inflammatory responses as well as the production and activation of various proteases promote aortic aneurysm, the specific mechanisms remain unclear. Researchers are further exploring the pathogenesis of aneurysms to find new targets for diagnosis and treatment. To better understand aortic aneurysm, this review elaborates on the discovery history of aortic aneurysm, main classification and clinical manifestations, related molecular mechanisms, clinical cohort studies and animal models, with the ultimate goal of providing insights into the treatment of this devastating disease. The underlying problem with aneurysm disease is weakening of the aortic wall, leading to progressive dilation. If not treated in time, the aortic aneurysm eventually ruptures. An aortic aneurysm is a local enlargement of an artery caused by a weakening of the aortic wall. The disease is usually asymptomatic but leads to high mortality due to the risk of artery rupture.
Collapse
|
7
|
Natural Bioactive Compounds Targeting NADPH Oxidase Pathway in Cardiovascular Diseases. Molecules 2023; 28:molecules28031047. [PMID: 36770715 PMCID: PMC9921542 DOI: 10.3390/molecules28031047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/04/2022] [Accepted: 12/10/2022] [Indexed: 01/21/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, in both developed and developing countries. According to the WHO report, the morbidity and mortality caused by CVD will continue to rise with the estimation of death going up to 22.2 million in 2030. NADPH oxidase (NOX)-derived reactive oxygen species (ROS) production induces endothelial nitric oxide synthase (eNOS) uncoupling and mitochondrial dysfunction, resulting in sustained oxidative stress and the development of cardiovascular diseases. Seven distinct members of the family have been identified of which four (namely, NOX1, 2, 4 and 5) may have cardiovascular functions. Currently, the treatment and management plan for patients with CVDs mainly depends on the drugs. However, prolonged use of prescribed drugs may cause adverse drug reactions. Therefore, it is crucial to find alternative treatment options with lesser adverse effects. Natural products have been gaining interest as complementary therapy for CVDs over the past decade due to their wide range of medicinal properties, including antioxidants. These might be due to their potent active ingredients, such as flavonoid and phenolic compounds. Numerous natural compounds have been demonstrated to have advantageous effects on cardiovascular disease via NADPH cascade. This review highlights the potential of natural products targeting NOX-derived ROS generation in treating CVDs. Emphasis is put on the activation of the oxidases, including upstream or downstream signalling events.
Collapse
|
8
|
NOX as a Therapeutic Target in Liver Disease. Antioxidants (Basel) 2022; 11:antiox11102038. [PMID: 36290761 PMCID: PMC9598239 DOI: 10.3390/antiox11102038] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022] Open
Abstract
The nicotinamide adenine dinucleotide phosphate hydrogen oxidase (NADPH oxidase or NOX) plays a critical role in the inflammatory response and fibrosis in several organs such as the lungs, pancreas, kidney, liver, and heart. In the liver, NOXs contribute, through the generation of reactive oxygen species (ROS), to hepatic fibrosis by acting through multiple pathways, including hepatic stellate cell activation, proliferation, survival, and migration of hepatic stellate cells; hepatocyte apoptosis, enhancement of fibrogenic mediators, and mediation of an inflammatory cascade in both Kupffer cells and hepatic stellate cells. ROS are overwhelmingly produced during malignant transformation and hepatic carcinogenesis (HCC), creating an oxidative microenvironment that can cause different and various types of cellular stress, including DNA damage, ER stress, cell death of damaged hepatocytes, and oxidative stress. NOX1, NOX2, and NOX4, members of the NADPH oxidase family, have been linked to the production of ROS in the liver. This review will analyze some diseases related to an increase in oxidative stress and its relationship with the NOX family, as well as discuss some therapies proposed to slow down or control the disease's progression.
Collapse
|
9
|
Miyano K, Okamoto S, Kajikawa M, Kiyohara T, Kawai C, Yamauchi A, Kuribayashi F. Regulation of Derlin-1-mediated degradation of NADPH oxidase partner p22 phox by thiol modification. Redox Biol 2022; 56:102479. [PMID: 36122532 PMCID: PMC9486109 DOI: 10.1016/j.redox.2022.102479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022] Open
Abstract
The transmembrane protein p22phox heterodimerizes with NADPH oxidase (Nox) 1–4 and is essential for the reactive oxygen species-producing capacity of oxidases. Missense mutations in the p22phox gene prevent the formation of phagocytic Nox2-based oxidase, which contributes to host defense. This results in chronic granulomatous disease (CGD), a severe primary immunodeficiency syndrome. In this study, we characterized missense mutations in p22phox (L51Q, L52P, E53V, and P55R) in the A22° type (wherein the p22phox protein is undetectable) of CGD. We demonstrated that these substitutions enhanced the degradation of the p22phox protein in the endoplasmic reticulum (ER) and the binding of p22phox to Derlin-1, a key component of ER-associated degradation (ERAD). Therefore, the L51-L52-E53-P55 sequence is responsible for protein stability in the ER. We observed that the oxidation of the thiol group of Cys-50, which is adjacent to the L51-L52-E53-P55 sequence, suppressed p22phox degradation. However, the suppression effect was markedly attenuated by the serine substitution of Cys-50. Blocking the free thiol of Cys-50 by alkylation or C50S substitution promoted the association of p22phox with Derlin-1. Derlin-1 depletion partially suppressed the degradation of p22phox mutant proteins. Furthermore, heterodimerization with p22phox (C50S) induced rapid degradation of not only Nox2 but also nonphagocytic Nox4 protein, which is responsible for redox signaling. Thus, the redox-sensitive Cys-50 appears to determine whether p22phox becomes a target for degradation by the ERAD system through its interaction with Derlin-1. Missense mutations in exon 3 of p22phox enhance the binding of p22phox to Derlin-1. Oxidation of the thiol group of p22phox Cys50 suppresses p22phox degradation. Serine substitution of Cys-50 increases the affinity of p22phox for Derlin-1. Stability of the p22phox protein is regulated by redox-sensitive Cys-50.
Collapse
Affiliation(s)
- Kei Miyano
- Department of Natural Sciences, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan; Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan.
| | - Shuichiro Okamoto
- Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan
| | - Mizuho Kajikawa
- Laboratory of Microbiology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Takuya Kiyohara
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Chikage Kawai
- Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan
| | - Akira Yamauchi
- Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan
| | - Futoshi Kuribayashi
- Department of Biochemistry, Kawasaki Medical School, 577 Matsushima Kurashiki, Okayama, 701-0192, Japan
| |
Collapse
|
10
|
Hahner F, Moll F, Warwick T, Hebchen DM, Buchmann GK, Epah J, Abplanalp W, Schader T, Günther S, Gilsbach R, Brandes RP, Schröder K. Nox4 promotes endothelial differentiation through chromatin remodeling. Redox Biol 2022; 55:102381. [PMID: 35810713 PMCID: PMC9287364 DOI: 10.1016/j.redox.2022.102381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/20/2022] [Indexed: 01/09/2023] Open
Abstract
RATIONALE Nox4 is a constitutively active NADPH oxidase that constantly produces low levels of H2O2. Thereby, Nox4 contributes to cell homeostasis and long-term processes, such as differentiation. The high expression of Nox4 seen in endothelial cells contrasts with the low abundance of Nox4 in stem cells, which are accordingly characterized by low levels of H2O2. We hypothesize that Nox4 is a major contributor to endothelial differentiation, is induced during the process of differentiation, and facilitates homeostasis of the resulting endothelial cells. OBJECTIVE To determine the role of No×4 in differentiation of murine inducible pluripotent stem cells (miPSC) into endothelial cells (ECs). METHODS AND RESULTS miPSC, generated from mouse embryonic wildtype (WT) and Nox4-/- fibroblasts, were differentiated into endothelial cells (miPSC-EC) by stimulation with BMP4 and VEGF. During this process, Nox4 expression increased and knockout of Nox4 prolonged the abundance of pluripotency markers, while expression of endothelial markers was delayed in differentiating Nox4-depleted iPSCs. Eventually, angiogenic capacity of iPSC-ECs is reduced in Nox4 deficient cells, indicating that an absence of Nox4 diminishes stability of the reached phenotype. As an underlying mechanism, we identified JmjD3 as a redox target of Nox4. iPSC-ECs lacking Nox4 display a lower nuclear abundance of the histone demethylase JmjD3, resulting in an increased triple methylation of histone 3 (H3K27me3), which serves as a repressive mark for several genes involved in differentiation. CONCLUSIONS Nox4 promotes differentiation of miPSCs into ECs by oxidation of JmjD3 and subsequent demethylation of H3K27me3, which forced endothelial differentiation and stability.
Collapse
Affiliation(s)
- F Hahner
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - F Moll
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - T Warwick
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - D M Hebchen
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - G K Buchmann
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - J Epah
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - W Abplanalp
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - T Schader
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - S Günther
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - R Gilsbach
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - R P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - K Schröder
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany.
| |
Collapse
|
11
|
Eid SA, Savelieff MG, Eid AA, Feldman EL. Nox, Nox, Are You There? The Role of NADPH Oxidases in the Peripheral Nervous System. Antioxid Redox Signal 2022; 37:613-630. [PMID: 34861780 PMCID: PMC9634986 DOI: 10.1089/ars.2021.0135] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 02/06/2023]
Abstract
Significance: Reactive oxygen species (ROS) contribute to multiple aspects of peripheral nervous system (PNS) biology ranging from physiological processes (e.g., axonal outgrowth and regeneration) to pathophysiology (e.g., nerve degeneration). Although ROS are derived from multiple sources, NADPH oxidase (Nox) family members are dedicated to ROS generation. Noxs are expressed in the PNS, and their overexpression is associated with detrimental effects on nerve function and contributes, at least in part, to peripheral neuropathies. Recent Advances: Of the seven members, studies mostly focused on Nox1, Nox2, and Nox4, which are expressed in the PNS in a cell-specific manner. We have also recently identified human Nox5 in sural nerve biopsies. When maintained at homeostatic levels, Noxs regulate several aspects of peripheral nerve health, most notably neurite outgrowth and axonal regeneration following nerve lesion. While Nox2 and Nox4 dysregulation is a major source of oxidative stress in PNS disorders, including neuropathic pain and diabetic peripheral neuropathy, recent evidence also implicates Nox1 and Nox5. Critical Issues: Although there is compelling evidence for a direct role of Noxs on nerve function, little is known about their subcellular localization, intercellular regulation, and interaction. These, together with redox signaling, are considered crucial components of nerve redox status. In addition, the lack of isoform-specific inhibitors limits conclusions about the physiological role of Noxs in the PNS and their therapeutic potential in peripheral neuropathies. Future Directions: Future research using isoform-specific genetic and pharmacological approaches are therefore needed to better understand the significance of Nox enzymes in PNS (patho) physiology. Antioxid. Redox Signal. 37, 613-630.
Collapse
Affiliation(s)
- Stéphanie A. Eid
- Department of Neurology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurology, NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Masha G. Savelieff
- Department of Neurology, NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| | - Assaad A. Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Eva L. Feldman
- Department of Neurology, School of Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Neurology, NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Tan R, Cong T, Xu G, Hao Z, Liao J, Xie Y, Lin Y, Yang X, Li Q, Liu Y, Xia YL. Anthracycline-Induced Atrial Structural and Electrical Remodeling Characterizes Early Cardiotoxicity and Contributes to Atrial Conductive Instability and Dysfunction. Antioxid Redox Signal 2022; 37:19-39. [PMID: 35081742 DOI: 10.1089/ars.2021.0002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aims: Cancer patients treated with anthracyclines are susceptible to atrial fibrillation (AF), while the mechanisms remain unclear. Due to sudden and unpredictable features, prediction of anthracycline-induced AF at early phase is difficult. Clinically, we tested whether anthracycline-induced early atrial remodeling in patients could be detected by echocardiography. Experimentally, we investigated the mechanisms of doxorubicin-induced atrial remodeling and AF in mice, and the protective effects of dexrazoxane and antioxidants. Methods and Results: Postsurgery breast cancer patients with an anthracycline-containing or anthracycline exclusion regimen were recruited for echocardiography before chemotherapy, and 3 and 6 months after chemotherapy. Mice were injected with doxorubicin or vehicle (5 mg/kg/week, 4 weeks), and left atrial diameter, electrical transmission, and AF inducibility were measured. Meanwhile, the level of reactive oxygen species (ROS), activity of antioxidant enzymes, cardiomyocyte size, vacuolization, inflammation, and fibrosis were also measured in mouse atria. The therapeutic effects of dexrazoxane and antioxidants on doxorubicin-induced changes in the aforementioned parameters were also determined. While ventricular parameters and functions were unchanged in cancer patients receiving anthracyclines before and after chemotherapy, left atrial reservoir and conduit function were decreased at 3 months postchemotherapy versus prechemotherapy. Doxorubicin-induced susceptibility to AF occurred in mice before onset of ventricular dysfunction. Doxorubicin-induced AF was via inducing structural remodeling (cardiomyocyte death, hypotrophy, and vacuolization) and electrical remodeling (reduction and redistribution of connexin 43) in atria, which was effectively prevented by dexrazoxane or antioxidants through inhibiting ROS generation or enhancing ROS elimination. Innovation and Conclusion: AF inducibility was induced after doxorubicin injection, which can be inhibited by repressing the ROS level. Antioxid. Redox Signal. 37, 19-39. The Clinical Trial Registration number is PJ-KS-KY-2019-73.
Collapse
Affiliation(s)
- Ruopeng Tan
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tao Cong
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guiwen Xu
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhujing Hao
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiawei Liao
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunpeng Xie
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yajuan Lin
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaolei Yang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingsong Li
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yang Liu
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yun-Long Xia
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
13
|
Sun X, Xiao H, Li S, Chen R, Lin Z, Yang Y, Chen Z, Deng L, Huang H. Connexin32 ameliorates epithelial-to-mesenchymal-transition in diabetic renal tubular via inhibiting NOX4. Pharmacol Res 2022; 176:106084. [PMID: 35051590 DOI: 10.1016/j.phrs.2022.106084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/31/2021] [Accepted: 01/11/2022] [Indexed: 12/22/2022]
Abstract
Renal tubulointerstitial fibrosis (RIF), characterized by epithelial-to-mesenchymal transition (EMT) of renal tubular epithelial cells (TECs), is the main cause of diabetic renal fibrosis. Oxidative stress plays a pivotal role in the development of diabetic RIF. Connexin32 (Cx32), prominently expressed in renal TECs, has emerged as an important player in the regulation of oxidative stress. However, the role of Cx32 in diabetic RIF has not been explored yet. Here, we showed that adenovirus-mediated Cx32 overexpression suppressed EMT to ameliorate RIF and renal function in STZ-induced diabetic mice, while knockout (KO) of Cx32 exacerbated RIF in diabetic mice. Moreover, overexpression of Cx32 inhibited EMT and the production of extra cellular matrix (ECM) in high glucose (HG) induced NRK-52E cells, whereas knockdown of Cx32 showed the opposite effects. Furthermore, we showed that NOX4, the main source of ROS in renal tubular, was down-regulated by Cx32. Mechanistically, Cx32 down-regulated the expression of PKC alpha in a carboxyl-terminal-dependent manner, thereby inhibiting the phosphorylation at Thr147 of p22phox triggered by PKC alpha, which ultimately repressed the formation of the p22phox-NOX4 complex to reduce the protein level of NOX4. Thus, we establish Cx32 as a novel target and confirm the protection mechanism in RIF.
Collapse
Affiliation(s)
- Xiaohong Sun
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Haiming Xiao
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shanshan Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Rui Chen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zeyuan Lin
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yan Yang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhiquan Chen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning 530021, China.
| | - Li Deng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China.
| | - Heqing Huang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
14
|
Villegas L, Nørremølle A, Freude K, Vilhardt F. Nicotinamide Adenine Dinucleotide Phosphate Oxidases Are Everywhere in Brain Disease, but Not in Huntington's Disease? Front Aging Neurosci 2021; 13:736734. [PMID: 34803655 PMCID: PMC8602359 DOI: 10.3389/fnagi.2021.736734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by neuronal loss and tissue atrophy mainly in the striatum and cortex. In the early stages of the disease, impairment of neuronal function, synaptic dysfunction and white matter loss precedes neuronal death itself. Relative to other neurodegenerative diseases such as Alzheimer's and Parkinson's disease and Amyotrophic Lateral Sclerosis, where the effects of either microglia or NADPH oxidases (NOXs) are recognized as important contributors to disease pathogenesis and progression, there is a pronounced lack of information in HD. This information void contrasts with evidence from human HD patients where blood monocytes and microglia are activated well before HD clinical symptoms (PET scans), and the clear signs of oxidative stress and inflammation in post mortem HD brain. Habitually, NOX activity and oxidative stress in the central nervous system (CNS) are equated with microglia, but research of the last two decades has carved out important roles for NOX enzyme function in neurons. Here, we will convey recent information about the function of NOX enzymes in neurons, and contemplate on putative roles of neuronal NOX in HD. We will focus on NOX-produced reactive oxygen species (ROS) as redox signaling molecules in/among neurons, and the specific roles of NOXs in important processes such as neurogenesis and lineage specification, neurite outgrowth and growth cone dynamics, and synaptic plasticity where NMDAR-dependent signaling, and long-term depression/potentiation are redox-regulated phenomena. HD animal models and induced pluripotent stem cell (iPSC) studies have made it clear that the very same physiological processes are also affected in HD, and we will speculate on possible roles for NOX in the pathogenesis and development of disease. Finally, we also take into account the limited information on microglia in HD and relate this to any contribution of NOX enzymes.
Collapse
Affiliation(s)
- Luisana Villegas
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne Nørremølle
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Herranz-Itúrbide M, Peñuelas-Haro I, Espinosa-Sotelo R, Bertran E, Fabregat I. The TGF-β/NADPH Oxidases Axis in the Regulation of Liver Cell Biology in Health and Disease. Cells 2021; 10:cells10092312. [PMID: 34571961 PMCID: PMC8470857 DOI: 10.3390/cells10092312] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 12/28/2022] Open
Abstract
The Transforming Growth Factor-beta (TGF-β) pathway plays essential roles in liver development and homeostasis and become a relevant factor involved in different liver pathologies, particularly fibrosis and cancer. The family of NADPH oxidases (NOXs) has emerged in recent years as targets of the TGF-β pathway mediating many of its effects on hepatocytes, stellate cells and macrophages. This review focuses on how the axis TGF-β/NOXs may regulate the biology of different liver cells and how this influences physiological situations, such as liver regeneration, and pathological circumstances, such as liver fibrosis and cancer. Finally, we discuss whether NOX inhibitors may be considered as potential therapeutic tools in liver diseases.
Collapse
Affiliation(s)
- Macarena Herranz-Itúrbide
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (M.H.-I.); (I.P.-H.); (R.E.-S.); (E.B.)
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Irene Peñuelas-Haro
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (M.H.-I.); (I.P.-H.); (R.E.-S.); (E.B.)
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rut Espinosa-Sotelo
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (M.H.-I.); (I.P.-H.); (R.E.-S.); (E.B.)
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Esther Bertran
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (M.H.-I.); (I.P.-H.); (R.E.-S.); (E.B.)
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 08908 Barcelona, Spain; (M.H.-I.); (I.P.-H.); (R.E.-S.); (E.B.)
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, 08907 Barcelona, Spain
- Correspondence: ; Tel.: +34-932-607-828
| |
Collapse
|
16
|
Oxidative Stress Links Aging-Associated Cardiovascular Diseases and Prostatic Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5896136. [PMID: 34336107 PMCID: PMC8313344 DOI: 10.1155/2021/5896136] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/17/2021] [Accepted: 07/03/2021] [Indexed: 11/29/2022]
Abstract
The incidence of chronic aging-associated diseases, especially cardiovascular and prostatic diseases, is increasing with the aging of society. Evidence indicates that cardiovascular diseases usually coexist with prostatic diseases or increase its risk, while the pathological mechanisms of these diseases are unknown. Oxidative stress plays an important role in the development of both cardiovascular and prostatic diseases. The levels of oxidative stress biomarkers are higher in patients with cardiovascular diseases, and these also contribute to the development of prostatic diseases, suggesting cardiovascular diseases may increase the risk of prostatic diseases via oxidative stress. This review summarizes the role of oxidative stress in cardiovascular and prostatic diseases and also focuses on the main shared pathways underlying these diseases, in order to provide potential prevention and treatment targets.
Collapse
|
17
|
From Mitochondria to Atherosclerosis: The Inflammation Path. Biomedicines 2021; 9:biomedicines9030258. [PMID: 33807807 PMCID: PMC8000234 DOI: 10.3390/biomedicines9030258] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation is a key process in metazoan organisms due to its relevance for innate defense against infections and tissue damage. However, inflammation is also implicated in pathological processes such as atherosclerosis. Atherosclerosis is a chronic inflammatory disease of the arterial wall where unstable atherosclerotic plaque rupture causing platelet aggregation and thrombosis may compromise the arterial lumen, leading to acute or chronic ischemic syndromes. In this review, we will focus on the role of mitochondria in atherosclerosis while keeping inflammation as a link. Mitochondria are the main source of cellular energy. Under stress, mitochondria are also capable of controlling inflammation through the production of reactive oxygen species (ROS) and the release of mitochondrial components, such as mitochondrial DNA (mtDNA), into the cytoplasm or into the extracellular matrix, where they act as danger signals when recognized by innate immune receptors. Primary or secondary mitochondrial dysfunctions are associated with the initiation and progression of atherosclerosis by elevating the production of ROS, altering mitochondrial dynamics and energy supply, as well as promoting inflammation. Knowing and understanding the pathways behind mitochondrial-based inflammation in atheroma progression is essential to discovering alternative or complementary treatments.
Collapse
|
18
|
Pagano PJ, Cifuentes-Pagano E. The Enigmatic Vascular NOX: From Artifact to Double Agent of Change: Arthur C. Corcoran Memorial Lecture - 2019. Hypertension 2021; 77:275-283. [PMID: 33390049 DOI: 10.1161/hypertensionaha.120.13897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NOXs (NADPH oxidases) comprise a family of proteins whose primary function is the production of reactive oxygen species, namely, superoxide anion and hydrogen peroxide. The prototype first being discovered and characterized in neutrophils, multiple NOXs are now known to be broadly expressed in cell and organ systems and whose phylogeny spans countless life forms beginning with prokaryotes. This long-enduring evolutionary conservation underscores the importance of fundamental NOX functions. This review chronicles a personal perspective of the field beginning with the discovery of NOXs in the vasculature and the advances achieved through the years as to our understanding of their mechanisms of action and role in oxidative stress and disease. Furthermore, applications of isoform-selective inhibitors to dissect the role of NOX isozymes in vascular biology, focusing on inflammation, pulmonary hypertension, and aging are described.
Collapse
Affiliation(s)
- Patrick J Pagano
- Department of Pharmacology and Chemical Biology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, PA
| |
Collapse
|
19
|
Novel and Converging Ways of NOX2 and SOD3 in Trafficking and Redox Signaling in Macrophages. Antioxidants (Basel) 2021; 10:antiox10020172. [PMID: 33503855 PMCID: PMC7911390 DOI: 10.3390/antiox10020172] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/16/2022] Open
Abstract
Macrophages and related tissue macrophage populations use the classical NADPH oxidase (NOX2) for the regulated production of superoxide and derived oxidants for pathogen combat and redox signaling. With an emphasis on macrophages, we discuss how sorting into secretory storage vesicles, agonist-responsive membrane trafficking, and segregation into sphingolipid and cholesterol-enriched microdomains (lipid rafts) determine the subcellular distribution and spatial organization of NOX2 and superoxide dismutase-3 (SOD3). We discuss how inflammatory activation of macrophages, in part through small GTPase Rab27A/B regulation of the secretory compartments, mediates the coalescence of these two proteins on the cell surface to deliver a focalized hydrogen peroxide output. In interplay with membrane-embedded oxidant transporters and redox sensitive target proteins, this arrangement allows for the autocrine and paracrine signaling, which govern macrophage activation states and transcriptional programs. By discussing examples of autocrine and paracrine redox signaling, we highlight why formation of spatiotemporal microenvironments where produced superoxide is rapidly converted to hydrogen peroxide and conveyed immediately to reach redox targets in proximal vicinity is required for efficient redox signaling. Finally, we discuss the recent discovery of macrophage-derived exosomes as vehicles of NOX2 holoenzyme export to other cells.
Collapse
|
20
|
Isaguliants M, Bayurova E, Avdoshina D, Kondrashova A, Chiodi F, Palefsky JM. Oncogenic Effects of HIV-1 Proteins, Mechanisms Behind. Cancers (Basel) 2021; 13:305. [PMID: 33467638 PMCID: PMC7830613 DOI: 10.3390/cancers13020305] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/28/2020] [Accepted: 01/04/2021] [Indexed: 02/08/2023] Open
Abstract
People living with human immunodeficiency virus (HIV-1) are at increased risk of developing cancer, such as Kaposi sarcoma (KS), non-Hodgkin lymphoma (NHL), cervical cancer, and other cancers associated with chronic viral infections. Traditionally, this is linked to HIV-1-induced immune suppression with depletion of CD4+ T-helper cells, exhaustion of lymphopoiesis and lymphocyte dysfunction. However, the long-term successful implementation of antiretroviral therapy (ART) with an early start did not preclude the oncological complications, implying that HIV-1 and its antigens are directly involved in carcinogenesis and may exert their effects on the background of restored immune system even when present at extremely low levels. Experimental data indicate that HIV-1 virions and single viral antigens can enter a wide variety of cells, including epithelial. This review is focused on the effects of five viral proteins: envelope protein gp120, accessory protein negative factor Nef, matrix protein p17, transactivator of transcription Tat and reverse transcriptase RT. Gp120, Nef, p17, Tat, and RT cause oxidative stress, can be released from HIV-1-infected cells and are oncogenic. All five are in a position to affect "innocent" bystander cells, specifically, to cause the propagation of (pre)existing malignant and malignant transformation of normal epithelial cells, giving grounds to the direct carcinogenic effects of HIV-1.
Collapse
Affiliation(s)
- Maria Isaguliants
- Gamaleya Research Center for Epidemiology and Microbiology, 123098 Moscow, Russia; (E.B.); (D.A.)
- M.P. Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia;
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden;
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia
| | - Ekaterina Bayurova
- Gamaleya Research Center for Epidemiology and Microbiology, 123098 Moscow, Russia; (E.B.); (D.A.)
- M.P. Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia;
| | - Darya Avdoshina
- Gamaleya Research Center for Epidemiology and Microbiology, 123098 Moscow, Russia; (E.B.); (D.A.)
- M.P. Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia;
| | - Alla Kondrashova
- M.P. Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia;
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Joel M. Palefsky
- Department of Medicine, University of California, San Francisco, CA 94117, USA;
| |
Collapse
|
21
|
Vermot A, Petit-Härtlein I, Breyton C, Le Roy A, Thépaut M, Vivès C, Moulin M, Härtlein M, Grudinin S, Smith SME, Ebel C, Martel A, Fieschi F. Interdomain Flexibility within NADPH Oxidase Suggested by SANS Using LMNG Stealth Carrier. Biophys J 2020; 119:605-618. [PMID: 32668232 PMCID: PMC7399496 DOI: 10.1016/j.bpj.2020.06.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/26/2020] [Accepted: 06/23/2020] [Indexed: 11/12/2022] Open
Abstract
Small angle neutron scattering (SANS) provides a method to obtain important low-resolution information for integral membrane proteins (IMPs), challenging targets for structural determination. Specific deuteration furnishes a "stealth" carrier for the solubilized IMP. We used SANS to determine a structural envelope of SpNOX, the Streptococcus pneumoniae NADPH oxidase (NOX), a prokaryotic model system for exploring structure and function of eukaryotic NOXes. SpNOX was solubilized in the detergent lauryl maltose neopentyl glycol, which provides optimal SpNOX stability and activity. Using deuterated solvent and protein, the lauryl maltose neopentyl glycol was experimentally undetected in SANS. This affords a cost-effective SANS approach for obtaining novel structural information on IMPs. Combining SANS data with molecular modeling provided a first, to our knowledge, structural characterization of an entire NOX enzyme. It revealed a distinctly less compact structure than that predicted from the docking of homologous crystal structures of the separate transmembrane and dehydrogenase domains, consistent with a flexible linker connecting the two domains.
Collapse
Affiliation(s)
- Annelise Vermot
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | | | - Cécile Breyton
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Aline Le Roy
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Michel Thépaut
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Corinne Vivès
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | | | | | | | - Susan M E Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, Georgia
| | - Christine Ebel
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | | | - Franck Fieschi
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France.
| |
Collapse
|
22
|
Oxidation of HDAC4 by Nox4-derived H 2O 2 maintains tube formation by endothelial cells. Redox Biol 2020; 36:101669. [PMID: 32818796 PMCID: PMC7452117 DOI: 10.1016/j.redox.2020.101669] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/29/2022] Open
Abstract
NADPH oxidases produce reactive oxygen species that differ in localization, type and concentration. Within the Nox family only Nox4 produces H2O2 which can directly oxidize cysteine residues. With this post-translational modification, activity, stability, localization and protein-protein interactions of the affected protein is altered. Nox4 controls differentiation, cellular homeostasis and prevents inflammation. Therefore, is likely that epigenetic mechanisms contribute to the effects of Nox4. One group of epigenetic modifiers are class IIa histone deacetylases (HDACs). We hypothesize that Nox4-derived H2O2 oxidizes HDACs and analyzed whether HDACs can be differentially oxidized by Nox4. As an artificial system, we utilized HEK293 cells, overexpressing Nox4 in a tetracycline-inducible manner. HDAC4 was oxidized upon Nox4 overexpression. Additionally, Nox4 overexpression increased HDAC4 phosphorylation on Ser632. H2O2 disrupted HDAC4/Mef2A complex, which de-represses Mef2A. In endothelial cells such as HUVECs and HMECs, overexpression of HDAC4 significantly reduced tube formation. Overexpression of a redox insensitive HDAC4 had no effect on endothelial tube formation. Treatment with H2O2, induction of Nox4 expression by treatment of the cells with TGFβ and co-overexpression of Nox4 not only induced phosphorylation of HDAC4, but also restored the repressive effect of HDAC4 for tube formation, while overexpression of a redox dead mutant of Nox4 did not. Taken together, Nox4 oxidizes HDAC4, increases its phosphorylation, and eventually ensures proper tube formation by endothelial cells.
Collapse
|
23
|
Negre-Salvayre A, Guerby P, Gayral S, Laffargue M, Salvayre R. Role of reactive oxygen species in atherosclerosis: Lessons from murine genetic models. Free Radic Biol Med 2020; 149:8-22. [PMID: 31669759 DOI: 10.1016/j.freeradbiomed.2019.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
Atherosclerosis is a multifactorial chronic and inflammatory disease of medium and large arteries, and the major cause of cardiovascular morbidity and mortality worldwide. The pathogenesis of atherosclerosis involves a number of risk factors and complex events including hypercholesterolemia, endothelial dysfunction, increased permeability to low density lipoproteins (LDL) and their sequestration on extracellular matrix in the intima of lesion-prone areas. These events promote LDL modifications, particularly by oxidation, which generates acute and chronic inflammatory responses implicated in atherogenesis and lesion progression. Reactive oxygen species (ROS) (which include both free radical and non-free radical oxygen intermediates), play a key-role at each step of atherogenesis, in endothelial dysfunction, LDL oxidation, and inflammatory events involved in the initiation and development of atherosclerosis lesions. Most advanced knowledge supporting the "oxidative theory of atherosclerosis" i.e. the nature and the cellular sources of ROS and antioxidant defences, as well as the mechanisms involved in the redox balance, is based on the use of genetically engineered animals, i.e. transgenic, genetically modified, or altered for systems producing or neutralizing ROS in the vessels. This review summarizes the results obtained from animals genetically manipulated for various sources of ROS or antioxidant defences in the vascular wall, and their relevance (advance or limitation), for understanding the place and role of ROS in atherosclerosis.
Collapse
Affiliation(s)
| | - Paul Guerby
- Inserm U-1048, Université de Toulouse, France; Pôle de gynécologie obstétrique, Hôpital Paule-de-Viguier, CHU de Toulouse, France
| | | | | | | |
Collapse
|
24
|
NADPH oxidases and oxidase crosstalk in cardiovascular diseases: novel therapeutic targets. Nat Rev Cardiol 2019; 17:170-194. [PMID: 31591535 DOI: 10.1038/s41569-019-0260-8] [Citation(s) in RCA: 326] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS)-dependent production of ROS underlies sustained oxidative stress, which has been implicated in the pathogenesis of cardiovascular diseases such as hypertension, aortic aneurysm, hypercholesterolaemia, atherosclerosis, diabetic vascular complications, cardiac ischaemia-reperfusion injury, myocardial infarction, heart failure and cardiac arrhythmias. Interactions between different oxidases or oxidase systems have been intensively investigated for their roles in inducing sustained oxidative stress. In this Review, we discuss the latest data on the pathobiology of each oxidase component, the complex crosstalk between different oxidase components and the consequences of this crosstalk in mediating cardiovascular disease processes, focusing on the central role of particular NADPH oxidase (NOX) isoforms that are activated in specific cardiovascular diseases. An improved understanding of these mechanisms might facilitate the development of novel therapeutic agents targeting these oxidase systems and their interactions, which could be effective in the prevention and treatment of cardiovascular disorders.
Collapse
|
25
|
Meitzler JL, Konaté MM, Doroshow JH. Hydrogen peroxide-producing NADPH oxidases and the promotion of migratory phenotypes in cancer. Arch Biochem Biophys 2019; 675:108076. [PMID: 31415727 DOI: 10.1016/j.abb.2019.108076] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 01/03/2023]
Abstract
The cellular microenvironment plays a critical role in cancer initiation and progression. Exposure to oxidative stress, specifically hydrogen peroxide (H2O2), has been linked to aberrant cellular signaling through which the development of cancer may be promoted. Three members of the NADPH oxidase family (NOX4, DUOX1 and DUOX2) explicitly generate this non-radical oxidant in a wide range of tissues, often in support of the inflammatory response. This review summarizes the contributions of each H2O2-producing NOX to the invasive behaviors of tumors and/or the epithelial-mesenchymal transition (EMT) in cancer that plays an essential role in metastasis. Tissue localization in tumorigenesis is also highlighted, with patient-derived TCGA microarray data profiled across 31 cancer cohorts to provide a comprehensive guide to the relevance of NOX4/DUOX1/DUOX2 in cancer studies.
Collapse
Affiliation(s)
- Jennifer L Meitzler
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA.
| | - Mariam M Konaté
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - James H Doroshow
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA; Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
26
|
Reactive Oxygen Species Signaling Promotes Hypoxia-Inducible Factor 1α Stabilization in Sonic Hedgehog-Driven Cerebellar Progenitor Cell Proliferation. Mol Cell Biol 2019; 39:MCB.00268-18. [PMID: 30692272 DOI: 10.1128/mcb.00268-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 01/23/2019] [Indexed: 12/21/2022] Open
Abstract
Cerebellar development is a highly regulated process involving numerous factors acting with high specificity, both temporally and by location. Part of this process involves extensive proliferation of cerebellar granule neuron precursors (CGNPs) induced by Sonic Hedgehog (SHH) signaling, but downstream effectors of mitogenic signaling are still being elucidated. Using primary CGNP cultures, a well-established model for SHH-driven proliferation, we show that SHH-treated CGNPs feature high levels of hypoxia-inducible factor 1α (HIF1α), which is known to promote glycolysis, stemness, and angiogenesis. In CGNPs cultured under normoxic conditions, HIF1α is posttranslationally stabilized in a manner dependent upon reactive oxygen species (ROS) and NADPH oxidase (NOX), both of which are also upregulated in these cells. Inhibition of NOX activity resulted in HIF1α destabilization and reduced levels of cyclin D2, a marker of CGNP proliferation. As CGNPs are the putative cells of origin for the SHH subtype of medulloblastoma and aberrant SHH signaling is implicated in other neoplasms, these studies may also have future relevance in the context of cancer. Taken together, our findings suggest that a better understanding of nonhypoxic HIF1α stabilization through NOX-induced ROS generation can provide insights into normal cell proliferation in cerebellar development and SHH-driven cell proliferation in cancers with aberrant SHH signaling.
Collapse
|
27
|
Ding M, Fang QH, Cui YT, Shen QL, Liu Q, Wang PH, Yu DM, Li CJ. Liraglutide prevents β-cell apoptosis via inactivation of NOX2 and its related signaling pathway. J Diabetes Complications 2019; 33:267-277. [PMID: 30772113 DOI: 10.1016/j.jdiacomp.2018.12.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/20/2018] [Accepted: 12/26/2018] [Indexed: 01/01/2023]
Abstract
AIMS High glucose (HG)-induced pancreatic β-cell apoptosis may be a major contributor to the progression of diabetes mellitus (DM). NADPH oxidase (NOX2) has been considered a crucial regulator in β-cell apoptosis. This study was designed to evaluate the impact of GLP-1 receptor agonist (GLP-1Ra) liraglutide on pancreatic β-cell apoptosis in diabetes and the underlying mechanisms involved. METHODS The diabetic rat models induced by streptozotocin (STZ) and a high fat diet (HFD) received 12 weeks of liraglutide treatment. Hyperglycemic clamp test was carried out to evaluate β-cell function in vivo. Flow cytometry analysis was used to measure apoptosis rates in vitro. DCFH-DA method was used to detected ROS level in vivo and in vitro. RESULTS Liraglutide significantly improved islet function and morphology in diabetic rats and decreased cell apoptosis rates. Thr183/Thr185 p-JNK1/2 and NOX2 levels reduced in diabetic rats and HG-induced INS-1 cell following liraglutide treatment. In addition, liraglutide upregulated the phosphorylation of AMPKα (p-AMPKα), which prevented NOX2 activation and alleviated HG-induced β-cell apoptosis. CONCLUSION The p-AMPKα/NOX2/JNK1/2 pathway is essential for liraglutide to attenuate HG-induced β-cell apoptosis, which further proves that GLP-1Ras may become promising therapeutics for diabetes mellitus.
Collapse
Affiliation(s)
- Min Ding
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin 300070, PR China
| | - Qian-Hua Fang
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin 300070, PR China
| | - Yuan-Tao Cui
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin 300070, PR China
| | - Qi-Ling Shen
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin 300070, PR China
| | - Qian Liu
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin 300070, PR China
| | - Peng-Hua Wang
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin 300070, PR China
| | - De-Min Yu
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin 300070, PR China.
| | - Chun-Jun Li
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin 300070, PR China.
| |
Collapse
|
28
|
Schröder K. NADPH oxidase-derived reactive oxygen species: Dosis facit venenum. Exp Physiol 2019; 104:447-452. [PMID: 30737851 PMCID: PMC6593456 DOI: 10.1113/ep087125] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/07/2019] [Indexed: 01/24/2023]
Abstract
New Findings What is the topic of this review? Within this review, the role of reactive oxygen species in cellular homeostasis, physiology and pathophysiology is discussed.
What advances does it highlight? The review provides new concepts of how reactive oxygen species influence gene expression, energy consumption and other aspects of the life of a cell. Furthermore, a model is provided to illustrate how reactive oxygen species elicit specific oxidation of target molecules.
Abstract Reactive oxygen species (ROS) have a long history of bad reputation. They are needed and effective in host defense, but on the contrary may induce situations of oxidative stress. Besides that, within recent years several soft functions (functions that may occur and are not directly connected to an effect, but may influence signaling in an indirect manner) of NADPH oxidases have been discovered, which are slowly eroding the image of the solely dangerous ROS. NADPH oxidase‐derived ROS serve to ease or enable signal transduction and to maintain homeostasis. However, there is still an enormous lag in the knowledge concerning target proteins and how ROS can elicit specific signalling in different cells and tissues. The present review summarizes some important functions of Nox2 and Nox4. Furthermore, although highly speculative, a model is provided of how those NADPH oxidases might be able to oxidize target proteins in a specific way. Many concepts mentioned in this review represent my personal view and are supported only in part by published studies.
Collapse
Affiliation(s)
- Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany.,German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| |
Collapse
|
29
|
Abstract
SIGNIFICANCE Angiogenesis is the formation of new vessels that sprout from existing vessels. This process is highly complex and requires a coordinated shift of the endothelial phenotype from a quiescent cell in the vessel wall into a migrating or proliferating cell. Such change in the life of the endothelial cell is induced by a variety of factors such as hypoxia, metabolic changes, or cytokines. Recent Advances: Within the last years, it became clear that the cellular redox state and oxidation of signaling molecules or phosphatases are critical modulators in angiogenesis. CRITICAL ISSUES According to the wide variety of stimuli that induce angiogenesis, a complex signaling network is needed to support a coordinated response of the endothelial cell. Reactive oxygen species (ROS) now are second messengers that either directly oxidize a target molecule or initiate a cascade of redox sensitive steps that transmit the signal. Further Directions: For the understanding of redox signaling, it is essential to recognize and accept that ROS do not represent master regulators of angiogenetic processes. They rather modulate existing signal cascades. This review summarizes some current findings on redox signaling in angiogenesis.
Collapse
Affiliation(s)
- Katrin Schröder
- 1 Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany.,2 German Center for Cardiovascular Research (DZHK), Rhine-Main, Frankfurt, Germany
| |
Collapse
|
30
|
Abstract
Extracellular hydrogen peroxide is required for thyroperoxidase-mediated thyroid hormone synthesis in the follicular lumen of the thyroid gland. Among the NADPH oxidases, dual oxidases, DUOX1 and DUOX2, constitute a distinct subfamily initially identified as thyroid oxidases, based on their level of expression in the thyroid. Despite their high sequence similarity, the two isoforms present distinct regulations, tissue expression, and catalytic functions. Inactivating mutations in many of the genes involved in thyroid hormone synthesis cause thyroid dyshormonogenesis associated with iodide organification defect. This chapter provides an overview of the genetic alterations in DUOX2 and its maturation factor, DUOXA2, causing inherited severe hypothyroidism that clearly demonstrate the physiological implication of this oxidase in thyroid hormonogenesis. Mutations in the DUOX2 gene have been described in permanent but also in transient forms of congenital hypothyroidism. Moreover, accumulating evidence demonstrates that the high phenotypic variability associated with altered DUOX2 function is not directly related to the number of inactivated DUOX2 alleles, suggesting the existence of other pathophysiological factors. The presence of two DUOX isoforms and their corresponding maturation factors in the same organ could certainly constitute an efficient redundant mechanism to maintain sufficient H2O2 supply for iodide organification. Many of the reported DUOX2 missense variants have not been functionally characterized, their clinical impact in the observed phenotype remaining unresolved, especially in mild transient congenital hypothyroidism. DUOX2 function should be carefully evaluated using an in vitro assay wherein (1) DUOXA2 is co-expressed, (2) H2O2 production is activated, (3) and DUOX2 membrane expression is precisely analyzed.
Collapse
Affiliation(s)
- Xavier De Deken
- Faculté de Médecine, Université Libre de Bruxelles (ULB), Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Brussels, Belgium.
| | - Françoise Miot
- Faculté de Médecine, Université Libre de Bruxelles (ULB), Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Brussels, Belgium
| |
Collapse
|
31
|
Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 2019; 99:311-379. [PMID: 30379623 PMCID: PMC6442925 DOI: 10.1152/physrev.00036.2017] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/30/2018] [Accepted: 05/06/2018] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of sufficient NO levels to regulate the resistance of the blood vessels and hence the maintenance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concentration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreductases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the therapeutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.
Collapse
Affiliation(s)
- Jesús Tejero
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Protective Role of Polyphenols against Vascular Inflammation, Aging and Cardiovascular Disease. Nutrients 2018; 11:nu11010053. [PMID: 30597847 PMCID: PMC6357531 DOI: 10.3390/nu11010053] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 01/02/2023] Open
Abstract
Aging is a major risk factor in the development of chronic diseases affecting various tissues including the cardiovascular system, muscle and bones. Age-related diseases are a consequence of the accumulation of cellular damage and reduced activity of protective stress response pathways leading to low-grade systemic inflammation and oxidative stress. Both inflammation and oxidative stress are major contributors to cellular senescence, a process in which cells stop proliferating and become dysfunctional by secreting inflammatory molecules, reactive oxygen species (ROS) and extracellular matrix components that cause inflammation and senescence in the surrounding tissue. This process is known as the senescence associated secretory phenotype (SASP). Thus, accumulation of senescent cells over time promotes the development of age-related diseases, in part through the SASP. Polyphenols, rich in fruits and vegetables, possess antioxidant and anti-inflammatory activities associated with protective effects against major chronic diseases, such as cardiovascular disease (CVD). In this review, we discuss molecular mechanisms by which polyphenols improve anti-oxidant capacity, mitochondrial function and autophagy, while reducing oxidative stress, inflammation and cellular senescence in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs). We also discuss the therapeutic potential of polyphenols in reducing the effects of the SASP and the incidence of CVD.
Collapse
|
33
|
Carvalho C, Moreira PI. Oxidative Stress: A Major Player in Cerebrovascular Alterations Associated to Neurodegenerative Events. Front Physiol 2018; 9:806. [PMID: 30018565 PMCID: PMC6037979 DOI: 10.3389/fphys.2018.00806] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/08/2018] [Indexed: 12/19/2022] Open
Abstract
The brain is one of the most exquisite organs in the body with high metabolic demands, and requires a tight regulation of the surrounding environment. This tight control is exerted by the neurovascular unit (NVU) comprising different cell types, where endothelial cells play the commander-in-chief role. Thus, it is assumable that even slight perturbations in NVU might affect, in some cases irreversibly, brain homeostasis and health. In this line, recent findings support the two-hit vascular hypothesis for neurodegenerative conditions, where vascular dysfunction underlies the development of neurodegenerative diseases, such as Alzheimer’s disease (AD). Knowing that endothelial cells are rich in mitochondria and nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, two major reactive oxygen species (ROS) sources, this review aims to gather information on how oxidative stress is in the front line of vascular alterations observed in brain aging and neurodegenerative conditions, particularly AD. Also, a brief discussion about the therapeutic strategies aimed to protect against cerebrovascular diseases is included.
Collapse
Affiliation(s)
- Cristina Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
34
|
Shih YC, Chen CL, Zhang Y, Mellor RL, Kanter EM, Fang Y, Wang HC, Hung CT, Nong JY, Chen HJ, Lee TH, Tseng YS, Chen CN, Wu CC, Lin SL, Yamada KA, Nerbonne JM, Yang KC. Endoplasmic Reticulum Protein TXNDC5 Augments Myocardial Fibrosis by Facilitating Extracellular Matrix Protein Folding and Redox-Sensitive Cardiac Fibroblast Activation. Circ Res 2018. [PMID: 29535165 DOI: 10.1161/circresaha.117.312130] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
RATIONALE Cardiac fibrosis plays a critical role in the pathogenesis of heart failure. Excessive accumulation of extracellular matrix (ECM) resulting from cardiac fibrosis impairs cardiac contractile function and increases arrhythmogenicity. Current treatment options for cardiac fibrosis, however, are limited, and there is a clear need to identify novel mediators of cardiac fibrosis to facilitate the development of better therapeutics. Exploiting coexpression gene network analysis on RNA sequencing data from failing human heart, we identified TXNDC5 (thioredoxin domain containing 5), a cardiac fibroblast (CF)-enriched endoplasmic reticulum protein, as a potential novel mediator of cardiac fibrosis, and we completed experiments to test this hypothesis directly. OBJECTIVE The objective of this study was to determine the functional role of TXNDC5 in the pathogenesis of cardiac fibrosis. METHODS AND RESULTS RNA sequencing and Western blot analyses revealed that TXNDC5 mRNA and protein were highly upregulated in failing human left ventricles and in hypertrophied/failing mouse left ventricle. In addition, cardiac TXNDC5 mRNA expression levels were positively correlated with those of transcripts encoding transforming growth factor β1 and ECM proteins in vivo. TXNDC5 mRNA and protein were increased in human CF (hCF) under transforming growth factor β1 stimulation in vitro. Knockdown of TXNDC5 attenuated transforming growth factor β1-induced hCF activation and ECM protein upregulation independent of SMAD3 (SMAD family member 3), whereas increasing expression of TXNDC5 triggered hCF activation and proliferation and increased ECM protein production. Further experiments showed that TXNDC5, a protein disulfide isomerase, facilitated ECM protein folding and that depletion of TXNDC5 led to ECM protein misfolding and degradation in CF. In addition, TXNDC5 promotes hCF activation and proliferation by enhancing c-Jun N-terminal kinase activity via increased reactive oxygen species, derived from NAD(P)H oxidase 4. Transforming growth factor β1-induced TXNDC5 upregulation in hCF was dependent on endoplasmic reticulum stress and activating transcription factor 6-mediated transcriptional control. Targeted disruption of Txndc5 in mice (Txndc5-/-) revealed protective effects against isoproterenol-induced cardiac hypertrophy, reduced fibrosis (by ≈70%), and markedly improved left ventricle function; post-isoproterenol left ventricular ejection fraction was 59.1±1.5 versus 40.1±2.5 (P<0.001) in Txndc5-/- versus wild-type mice, respectively. CONCLUSIONS The endoplasmic reticulum protein TXNDC5 promotes cardiac fibrosis by facilitating ECM protein folding and CF activation via redox-sensitive c-Jun N-terminal kinase signaling. Loss of TXNDC5 protects against β agonist-induced cardiac fibrosis and contractile dysfunction. Targeting TXNDC5, therefore, could be a powerful new therapeutic approach to mitigate excessive cardiac fibrosis, thereby improving cardiac function and outcomes in patients with heart failure.
Collapse
Affiliation(s)
- Ying-Chun Shih
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Chao-Ling Chen
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Yan Zhang
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Rebecca L Mellor
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Evelyn M Kanter
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Yun Fang
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Hua-Chi Wang
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Chen-Ting Hung
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Jing-Yi Nong
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Hui-Ju Chen
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Tzu-Han Lee
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Yi-Shuan Tseng
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Chiung-Nien Chen
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Chau-Chung Wu
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Shuei-Liong Lin
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Kathryn A Yamada
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Jeanne M Nerbonne
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei
| | - Kai-Chien Yang
- From the Department and Graduate Institute of Pharmacology (Y.-C.S., C.-L.C., H.-C.W., C.-T.H., J.-Y.N., H.-J.C., T.-H.L., Y.-S.T., K.-C.Y.), Department and Graduate Institute of Medical Education and Bioethics (C.-C.W.), and Department and Graduate Institute of Physiology (S.-L.L.), National Taiwan University College of Medicine, Taipei; Department of Developmental Biology (J.M.N.) and Center for Cardiovascular Research, Cardiovascular Division, Department of Medicine (Y.Z., R.L.M., E.M.K., K.A.Y., J.M.N.), Washington University School of Medicine, St Louis, MO; Department of Medicine, University of Chicago, IL (Y.F.); Department of Surgery (C.-N.C.), Division of Nephrology, Department of Internal Medicine (S.-L.L.), and Division of Cardiology, Department of Internal Medicine (C.-C.W., K.-C.Y.), National Taiwan University Hospital, Taipei.
| |
Collapse
|
35
|
Moloney JN, Jayavelu AK, Stanicka J, Roche SL, O'Brien RL, Scholl S, Böhmer FD, Cotter TG. Nuclear membrane-localised NOX4D generates pro-survival ROS in FLT3-ITD-expressing AML. Oncotarget 2017; 8:105440-105457. [PMID: 29285262 PMCID: PMC5739649 DOI: 10.18632/oncotarget.22241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022] Open
Abstract
Internal tandem duplication of the juxtamembrane domain of FMS-like tyrosine kinase 3 (FLT3-ITD) is the most prevalent genetic aberration present in 20-30% of acute myeloid leukaemia (AML) cases and is associated with a poor prognosis. FLT3-ITD expressing cells express elevated levels of NADPH oxidase 4 (NOX4)-generated pro-survival hydrogen peroxide (H2O2) contributing to increased levels of DNA oxidation and double strand breaks. NOX4 is constitutively active and has been found to have various isoforms expressed at multiple locations within a cell. The purpose of this study was to investigate the expression, localisation and regulation of NOX4 28 kDa splice variant, NOX4D. NOX4D has previously been shown to localise to the nucleus and nucleolus in various cell types and is implicated in the generation of reactive oxygen species (ROS) and DNA damage. Here, we demonstrate that FLT3-ITD expressing-AML patient samples as well as -cell lines express the NOX4D isoform resulting in elevated H2O2 levels compared to FLT3-WT expressing cells, as quantified by flow cytometry. Cell fractionation indicated that NOX4D is nuclear membrane-localised in FLT3-ITD expressing cells. Treatment of MV4-11 cells with receptor trafficking inhibitors, tunicamycin and brefeldin A, resulted in deglycosylation of NOX4 and NOX4D. Inhibition of the FLT3 receptor revealed that the FLT3-ITD oncogene is responsible for the production of NOX4D-generated H2O2 in AML. We found that inhibition of the PI3K/AKT and STAT5 pathways resulted in down-regulation of NOX4D-generated pro-survival ROS. Taken together these findings indicate that nuclear membrane-localised NOX4D-generated pro-survival H2O2 may be contributing to genetic instability in FLT3-ITD expressing AML.
Collapse
Affiliation(s)
- Jennifer N Moloney
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Ashok Kumar Jayavelu
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany.,Current address: Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Joanna Stanicka
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Sarah L Roche
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Rebecca L O'Brien
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Sebastian Scholl
- Department of Haematology/Oncology, Clinic for Internal Medicine II, Jena University Hospital, Jena, Germany
| | - Frank-D Böhmer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Thomas G Cotter
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Meitzler JL, Makhlouf HR, Antony S, Wu Y, Butcher D, Jiang G, Juhasz A, Lu J, Dahan I, Jansen-Dürr P, Pircher H, Shah AM, Roy K, Doroshow JH. Decoding NADPH oxidase 4 expression in human tumors. Redox Biol 2017; 13:182-195. [PMID: 28578276 PMCID: PMC5458090 DOI: 10.1016/j.redox.2017.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 05/23/2017] [Indexed: 12/27/2022] Open
Abstract
NADPH oxidase 4 (NOX4) is a redox active, membrane-associated protein that contributes to genomic instability, redox signaling, and radiation sensitivity in human cancers based on its capacity to generate H2O2 constitutively. Most studies of NOX4 in malignancy have focused on the evaluation of a small number of tumor cell lines and not on human tumor specimens themselves; furthermore, these studies have often employed immunological tools that have not been well characterized. To determine the prevalence of NOX4 expression across a broad range of solid tumors, we developed a novel monoclonal antibody that recognizes a specific extracellular region of the human NOX4 protein, and that does not cross-react with any of the other six members of the NOX gene family. Evaluation of 20 sets of epithelial tumors revealed, for the first time, high levels of NOX4 expression in carcinomas of the head and neck (15/19 patients), esophagus (12/18 patients), bladder (10/19 patients), ovary (6/17 patients), and prostate (7/19 patients), as well as malignant melanoma (7/15 patients) when these tumors were compared to histologically-uninvolved specimens from the same organs. Detection of NOX4 protein upregulation by low levels of TGF-β1 demonstrated the sensitivity of this new probe; and immunofluorescence experiments found that high levels of endogenous NOX4 expression in ovarian cancer cells were only demonstrable associated with perinuclear membranes. These studies suggest that NOX4 expression is upregulated, compared to normal tissues, in a well-defined, and specific group of human carcinomas, and that its expression is localized on intracellular membranes in a fashion that could modulate oxidative DNA damage.
Collapse
Affiliation(s)
- Jennifer L Meitzler
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Hala R Makhlouf
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Smitha Antony
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yongzhong Wu
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Donna Butcher
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21702, USA
| | - Guojian Jiang
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Agnes Juhasz
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jiamo Lu
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Iris Dahan
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, 6020 Innsbruck, Austria
| | - Haymo Pircher
- Institute for Biomedical Aging Research and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, 6020 Innsbruck, Austria
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, Cardiovascular Division, James Black Centre, London SE5 9NU, United Kingdom
| | - Krishnendu Roy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - James H Doroshow
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
37
|
Little AC, Sulovari A, Danyal K, Heppner DE, Seward DJ, van der Vliet A. Paradoxical roles of dual oxidases in cancer biology. Free Radic Biol Med 2017; 110:117-132. [PMID: 28578013 PMCID: PMC5535817 DOI: 10.1016/j.freeradbiomed.2017.05.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023]
Abstract
Dysregulated oxidative metabolism is a well-recognized aspect of cancer biology, and many therapeutic strategies are based on targeting cancers by altering cellular redox pathways. The NADPH oxidases (NOXes) present an important enzymatic source of biological oxidants, and the expression and activation of several NOX isoforms are frequently dysregulated in many cancers. Cell-based studies have demonstrated a role for several NOX isozymes in controlling cell proliferation and/or cell migration, further supporting a potential contributing role for NOX in promoting cancer. While various NOX isoforms are often upregulated in cancers, paradoxical recent findings indicate that dual oxidases (DUOXes), normally prominently expressed in epithelial lineages, are frequently suppressed in epithelial-derived cancers by epigenetic mechanisms, although the functional relevance of such DUOX silencing has remained unclear. This review will briefly summarize our current understanding regarding the importance of reactive oxygen species (ROS) and NOXes in cancer biology, and focus on recent observations indicating the unique and seemingly opposing roles of DUOX enzymes in cancer biology. We will discuss current knowledge regarding the functional properties of DUOX, and recent studies highlighting mechanistic consequences of DUOX1 loss in lung cancer, and its consequences for tumor invasiveness and current anticancer therapy. Finally, we will also discuss potentially unique roles for the DUOX maturation factors. Overall, a better understanding of mechanisms that regulate DUOX and the functional consequences of DUOX silencing in cancer may offer valuable new diagnostic insights and novel therapeutic opportunities.
Collapse
Affiliation(s)
- Andrew C Little
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States
| | - Arvis Sulovari
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States; Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - Karamatullah Danyal
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - David E Heppner
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - David J Seward
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States.
| |
Collapse
|
38
|
Yu W, Ji W, Mi L, Lin C. Mechanisms of N‑acetylcysteine in reducing monocrotaline‑induced pulmonary hypertension in rats: Inhibiting the expression of Nox1 in pulmonary vascular smooth muscle cells. Mol Med Rep 2017; 16:6148-6155. [PMID: 28849167 DOI: 10.3892/mmr.2017.7326] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 05/30/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the impact of N‑acetylcysteine (NAC) on the expression of reduced nicotinamide adenine dinucleotide phosphate oxidase 1 (Nox1), and the proliferation and apoptosis of pulmonary artery smooth muscle cells (PASMCs) in rats exhibiting monocrotaline (MCT)‑induced pulmonary hypertension, and to investigate the possible mechanisms and treatment roles of NAC in pulmonary vascular remodeling (PVR). A total of 18 Wistar rats were randomly divided into three groups: The control (C) group; the MCT (M) group; and the NAC (N) group. The right ventricular hypertrophy index (RVHI) and other indicators were recorded 6 weeks subsequently. Groups C and M were divided into two subgroups: Groups C1 and M1 (control); and group C2 and M2 group (treated with ML171). Group N was not sub‑divided. PASMCs were isolated, and the vascular remodeling and Nox1 positioning were observed. The expression of Nox mRNA in each group, and the proliferation, apoptosis, and superoxide dismutase (SOD) activity of PASMCs, prior to and following the ML171 treatment, were measured. NAC was able to decrease RVHI and other indicators (P<0.001). The mRNA expression of Nox1 and Nox4 in group M was significantly increased compared with group C (P<0.05), and NAC was able to significantly decrease the expression of these two factors in lung tissue (P<0.001). MCT‑PASMCs exhibited differences in Nox1 mRNA expression (P<0.001), and the total SOD activity was Nox1‑dependently increased (r=0.949; P<0.001). NAC was able to decrease Nox1‑derived reactive oxygen species in PASMCs, thereby improving PVR. Nox1 was able to increase SOD activity, thereby demonstrating its positive effect on the proliferation of MCT‑PASMCs.
Collapse
Affiliation(s)
- Wencheng Yu
- Department of Respiratory Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Weina Ji
- Department of Respiratory Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Liyun Mi
- Department of Respiratory Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Chen Lin
- Department of Respiratory Diseases, Shenli Oilfield Central Hospital, Dongying, Liaoning 257064, P.R. China
| |
Collapse
|
39
|
l -Carnitine ameliorates the oxidative stress response to angiotensin II by modulating NADPH oxidase through a reduction in protein kinase c activity and NF-κB translocation to the nucleus. Food Chem 2017; 228:356-366. [DOI: 10.1016/j.foodchem.2017.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 10/20/2016] [Accepted: 02/02/2017] [Indexed: 01/04/2023]
|
40
|
Li Y, Pagano PJ. Microvascular NADPH oxidase in health and disease. Free Radic Biol Med 2017; 109:33-47. [PMID: 28274817 PMCID: PMC5482368 DOI: 10.1016/j.freeradbiomed.2017.02.049] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/24/2017] [Accepted: 02/28/2017] [Indexed: 02/07/2023]
Abstract
The systemic and cerebral microcirculation contribute critically to regulation of local and global blood flow and perfusion pressure. Microvascular dysfunction, commonly seen in numerous cardiovascular pathologies, is associated with alterations in the oxidative environment including potentiated production of reactive oxygen species (ROS) and subsequent activation of redox signaling pathways. NADPH oxidases (Noxs) are a primary source of ROS in the vascular system and play a central role in cardiovascular health and disease. In this review, we focus on the roles of Noxs in ROS generation in resistance arterioles and capillaries, and summarize their contributions to microvascular physiology and pathophysiology in both systemic and cerebral microcirculation. In light of the accumulating evidence that Noxs are pivotal players in vascular dysfunction of resistance arterioles, selectively targeting Nox isozymes could emerge as a novel and effective therapeutic strategy for preventing and treating microvascular diseases.
Collapse
Affiliation(s)
- Yao Li
- Department of Pharmacology & Chemical Biology, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Patrick J Pagano
- Department of Pharmacology & Chemical Biology, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
41
|
Chen Q, Wang Q, Zhu J, Xiao Q, Zhang L. Reactive oxygen species: key regulators in vascular health and diseases. Br J Pharmacol 2017; 175:1279-1292. [PMID: 28430357 DOI: 10.1111/bph.13828] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 03/31/2017] [Accepted: 04/11/2017] [Indexed: 12/18/2022] Open
Abstract
ROS are a group of small reactive molecules that play critical roles in the regulation of various cell functions and biological processes. In the vascular system, physiological levels of ROS are essential for normal vascular functions including endothelial homeostasis and smooth muscle cell contraction. In contrast, uncontrolled overproduction of ROS resulting from an imbalance of ROS generation and elimination leads to the development of vascular diseases. Excessive ROS cause vascular cell damage, the recruitment of inflammatory cells, lipid peroxidation, activation of metalloproteinases and deposition of extracellular matrix, collectively leading to vascular remodelling. Evidence from a large number of studies has revealed that ROS and oxidative stress are involved in the initiation and progression of numerous vascular diseases including hypertension, atherosclerosis, restenosis and abdominal aortic aneurysm. Furthermore, considerable research has been implemented to explore antioxidants that can reduce ROS production and oxidative stress in order to ameliorate vascular diseases. In this review, we will discuss the nature and sources of ROS, their roles in vascular homeostasis and specific vascular diseases and various antioxidants as well as some of the pharmacological agents that are capable of reducing ROS and oxidative stress. The aim of this review is to provide information for developing promising clinical strategies targeting ROS to decrease cardiovascular risks. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Qishan Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiwen Wang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianhua Zhu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
42
|
Ivanov AV, Valuev-Elliston VT, Ivanova ON, Kochetkov SN, Starodubova ES, Bartosch B, Isaguliants MG. Oxidative Stress during HIV Infection: Mechanisms and Consequences. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8910396. [PMID: 27829986 PMCID: PMC5088339 DOI: 10.1155/2016/8910396] [Citation(s) in RCA: 234] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/18/2016] [Indexed: 12/15/2022]
Abstract
It is generally acknowledged that reactive oxygen species (ROS) play crucial roles in a variety of natural processes in cells. If increased to levels which cannot be neutralized by the defense mechanisms, they damage biological molecules, alter their functions, and also act as signaling molecules thus generating a spectrum of pathologies. In this review, we summarize current data on oxidative stress markers associated with human immunodeficiency virus type-1 (HIV-1) infection, analyze mechanisms by which this virus triggers massive ROS production, and describe the status of various defense mechanisms of the infected host cell. In addition, we have scrutinized scarce data on the effect of ROS on HIV-1 replication. Finally, we present current state of knowledge on the redox alterations as crucial factors of HIV-1 pathogenicity, such as neurotoxicity and dementia, exhaustion of CD4+/CD8+ T-cells, predisposition to lung infections, and certain side effects of the antiretroviral therapy, and compare them to the pathologies associated with the nitrosative stress.
Collapse
Affiliation(s)
- Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Vladimir T. Valuev-Elliston
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Olga N. Ivanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Sergey N. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | - Elizaveta S. Starodubova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
- M. P. Chumakov Institute of Poliomyelitis and Viral Encephalitides, Moscow 142782, Russia
| | - Birke Bartosch
- Cancer Research Center Lyon, INSERM U1052 and CNRS 5286, Lyon University, 69003 Lyon, France
- DevWeCan Laboratories of Excellence Network (Labex), France
| | - Maria G. Isaguliants
- Riga Stradins University, Riga LV-1007, Latvia
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
- N. F. Gamaleya Research Center of Epidemiology and Microbiology, Moscow 123098, Russia
| |
Collapse
|
43
|
Marschall R, Siegmund U, Burbank J, Tudzynski P. Update on Nox function, site of action and regulation in Botrytis cinerea. Fungal Biol Biotechnol 2016; 3:8. [PMID: 28955467 PMCID: PMC5611593 DOI: 10.1186/s40694-016-0026-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/29/2016] [Indexed: 12/11/2022] Open
Abstract
Background The production of reactive oxygen species (ROS) and a balanced redox homeostasis are essential parameters, which control the infection process of the plant pathogen Botrytis cinerea. The necrotrophic fungus is able to cope with the plants’ oxidative burst and even produces its own ROS to overcome the plants’ defense barrier. Major enzyme complexes, which are responsible for the production of superoxide, are NADPH oxidase (Nox) complexes. They play a central role in various growth, differentiation and pathogenic processes. However, information about their regulation and the integration in the complex signaling network of filamentous fungi is still scarce. Results In this work, we give an update on Nox structure, function, site of action and regulation. We show that functionality of the catalytic Nox-subunits seems to be independent from their transcriptional regulation and that the membrane orientation of BcNoxA would allow electron transport inside the ER. Following previous studies, which provided evidence for distinct functions of the NoxA complex inside the ER, we highlight in this work that the N-terminus of BcNoxA is essential for these functions. Finally, we elucidate the role of BcNoxD and BcNoxB inside the ER by complementing the deletion mutants with ER bound alleles. Conclusions This study provides a deeper analysis of the Nox complexes in B. cinerea. Besides new insights in the overall regulation of the complexes, we provide further evidence that the NoxA complex has a predominant role inside the ER, while the NoxB complex is mainly important outside the ER, likely at the plasma membrane. By considering all other putative Nox complex members, we propose a putative model, which describes the distinct complex pattern upon certain differentiation processes. Electronic supplementary material The online version of this article (doi:10.1186/s40694-016-0026-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robert Marschall
- Institut für Biologie und Biotechnologie der Pflanzen, Westfälische Wilhelms Universität, Schlossplatz 8, 48143 Münster, Germany
| | - Ulrike Siegmund
- Institut für Biologie und Biotechnologie der Pflanzen, Westfälische Wilhelms Universität, Schlossplatz 8, 48143 Münster, Germany
| | - Joachim Burbank
- Institut für Biologie und Biotechnologie der Pflanzen, Westfälische Wilhelms Universität, Schlossplatz 8, 48143 Münster, Germany
| | - Paul Tudzynski
- Institut für Biologie und Biotechnologie der Pflanzen, Westfälische Wilhelms Universität, Schlossplatz 8, 48143 Münster, Germany
| |
Collapse
|
44
|
CRISPR/Cas9-mediated knockout of p22phox leads to loss of Nox1 and Nox4, but not Nox5 activity. Redox Biol 2016; 9:287-295. [PMID: 27614387 PMCID: PMC5021817 DOI: 10.1016/j.redox.2016.08.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 08/16/2016] [Indexed: 11/22/2022] Open
Abstract
The NADPH oxidases are important transmembrane proteins producing reactive oxygen species (ROS). Within the Nox family, different modes of activation can be discriminated. Nox1-3 are dependent on different cytosolic subunits, Nox4 seems to be constitutively active and Nox5 is directly activated by calcium. With the exception of Nox5, all Nox family members are thought to depend on the small transmembrane protein p22phox. With the discovery of the CRISPR/Cas9-system, a tool to alter genomic DNA sequences has become available. So far, this method has not been widely used in the redox community. On such basis, we decided to study the requirement of p22phox in the Nox complex using CRISPR/Cas9-mediated knockout. Knockout of the gene of p22phox, CYBA, led to an ablation of activity of Nox4 and Nox1 but not of Nox5. Production of hydrogen peroxide or superoxide after knockout could be rescued with either human or rat p22phox, but not with the DUOX-maturation factors DUOXA1/A2. Furthermore, different mutations of p22phox were studied regarding the influence on Nox4-dependent H2O2 production. P22phox Q130* and Y121H affected maturation and activity of Nox4. Hence, Nox5-dependent O2•- production is independent of p22phox, but native p22phox is needed for maturation of Nox4 and production of H2O2.
Collapse
|
45
|
Brandes RP, Harenkamp S, Schürmann C, Josipovic I, Rashid B, Rezende F, Löwe O, Moll F, Epah J, Eresch J, Nayak A, Kopaliani I, Penski C, Mittelbronn M, Weissmann N, Schröder K. The Cytosolic NADPH Oxidase Subunit NoxO1 Promotes an Endothelial Stalk Cell Phenotype. Arterioscler Thromb Vasc Biol 2016; 36:1558-65. [PMID: 27283741 DOI: 10.1161/atvbaha.116.307132] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 05/31/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Reactive oxygen species generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidases contribute to angiogenesis and vascular repair. NADPH oxidase organizer 1 (NoxO1) is a cytosolic protein facilitating assembly of constitutively active NADPH oxidases. We speculate that NoxO1 also contributes to basal reactive oxygen species formation in the vascular system and thus modulates angiogenesis. APPROACH AND RESULTS A NoxO1 knockout mouse was generated, and angiogenesis was studied in cultured cells and in vivo. Angiogenesis of the developing retina and after femoral artery ligation was increased in NoxO1(-/-) when compared with wild-type animals. Spheroid outgrowth assays revealed greater angiogenic capacity of NoxO1(-/-) lung endothelial cells (LECs) and a more tip-cell-like phenotype than wild-type LECs. Usually signaling by the Notch pathway switches endothelial cells from a tip into a stalk cell phenotype. NoxO1(-/-) LECs exhibited attenuated Notch signaling as a consequence of an attenuated release of the Notch intracellular domain on ligand stimulation. This release is mediated by proteolytic cleavage involving the α-secretase ADAM17. For maximal activity, ADAM17 has to be oxidized, and overexpression of NoxO1 promoted this mode of activation. Moreover, the activity of ADAM17 was reduced in NoxO1(-/-) LECs when compared with wild-type LECs. CONCLUSIONS NoxO1 stimulates α-secretase activity probably through reactive oxygen species-mediated oxidation. Deletion of NoxO1 attenuates Notch signaling and thereby promotes a tip-cell phenotype that results in increased angiogenesis.
Collapse
Affiliation(s)
- Ralf P Brandes
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Sabine Harenkamp
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Christoph Schürmann
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Ivana Josipovic
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Beliza Rashid
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Flavia Rezende
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Oliver Löwe
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Franziska Moll
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Jeremy Epah
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Jeanette Eresch
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Arnab Nayak
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Irakli Kopaliani
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Cornelia Penski
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Michel Mittelbronn
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Norbert Weissmann
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Katrin Schröder
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.).
| |
Collapse
|
46
|
Abstract
Reactive oxygen species (ROS) and oxidative stress have long been linked to aging and diseases prominent in the elderly such as hypertension, atherosclerosis, diabetes and atrial fibrillation (AF). NADPH oxidases (Nox) are a major source of ROS in the vasculature and are key players in mediating redox signalling under physiological and pathophysiological conditions. In this review, we focus on the Nox-mediated ROS signalling pathways involved in the regulation of 'longevity genes' and recapitulate their role in age-associated vascular changes and in the development of age-related cardiovascular diseases (CVDs). This review is predicated on burgeoning knowledge that Nox-derived ROS propagate tightly regulated yet varied signalling pathways, which, at the cellular level, may lead to diminished repair, the aging process and predisposition to CVDs. In addition, we briefly describe emerging Nox therapies and their potential in improving the health of the elderly population.
Collapse
|
47
|
Haslund-Vinding J, McBean G, Jaquet V, Vilhardt F. NADPH oxidases in oxidant production by microglia: activating receptors, pharmacology and association with disease. Br J Pharmacol 2016; 174:1733-1749. [PMID: 26750203 DOI: 10.1111/bph.13425] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 12/15/2015] [Accepted: 01/07/2016] [Indexed: 12/26/2022] Open
Abstract
Microglia are the resident immune cells of the CNS and constitute a self-sustaining population of CNS-adapted tissue macrophages. As mononuclear phagocytic cells, they express high levels of superoxide-producing NADPH oxidases (NOX). The sole function of the members of the NOX family is to generate reactive oxygen species (ROS) that are believed to be important in CNS host defence and in the redox signalling circuits that shape the different activation phenotypes of microglia. NOX are also important in pathological conditions, where over-generation of ROS contributes to neuronal loss via direct oxidative tissue damage or disruption of redox signalling circuits. In this review, we assess the evidence for involvement of NOX in CNS physiopathology, with particular emphasis on the most important surface receptors that lead to generation of NOX-derived ROS. We evaluate the potential significance of the subcellular distribution of NOX isoforms for redox signalling or release of ROS to the extracellular medium. Inhibitory mechanisms that have been reported to restrain NOX activity in microglia and macrophages in vivo are also discussed. We provide a critical appraisal of frequently used and recently developed NOX inhibitors. Finally, we review the recent literature on NOX and other sources of ROS that are involved in activation of the inflammasome and discuss the potential influence of microglia-derived oxidants on neurogenesis, neural differentiation and culling of surplus progenitor cells. The degree to which excessive, badly timed or misplaced NOX activation in microglia may affect neuronal homeostasis in physiological or pathological conditions certainly merits further investigation. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- J Haslund-Vinding
- Institute of Cellular and Molecular Medicine, Copenhagen University, Copenhagen, Denmark.,Department of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - G McBean
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Ireland
| | - V Jaquet
- Department of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - F Vilhardt
- Institute of Cellular and Molecular Medicine, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
48
|
Prior KK, Wittig I, Leisegang MS, Groenendyk J, Weissmann N, Michalak M, Jansen-Dürr P, Shah AM, Brandes RP. The Endoplasmic Reticulum Chaperone Calnexin Is a NADPH Oxidase NOX4 Interacting Protein. J Biol Chem 2016; 291:7045-59. [PMID: 26861875 PMCID: PMC4807287 DOI: 10.1074/jbc.m115.710772] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Indexed: 11/24/2022] Open
Abstract
Within the family of NADPH oxidases, NOX4 is unique as it is predominantly localized in the endoplasmic reticulum, has constitutive activity, and generates hydrogen peroxide (H2O2). We hypothesize that these features are consequences of a so far unidentified NOX4-interacting protein. Two-dimensional blue native (BN) electrophorese combined with SDS-PAGE yielded NOX4 to reside in macromolecular complexes. Interacting proteins were screened by quantitative SILAC (stable isotope labeling of amino acids in cell culture) co-immunoprecipitation (Co-IP) in HEK293 cells stably overexpressing NOX4. By this technique, several interacting proteins were identified with calnexin showing the most robust interaction. Calnexin also resided in NOX4-containing complexes as demonstrated by complexome profiling from BN-PAGE. The calnexin NOX4 interaction could be confirmed by reverse Co-IP and proximity ligation assay, whereas NOX1, NOX2, or NOX5 did not interact with calnexin. Calnexin deficiency as studied in mouse embryonic fibroblasts from calnexin−/− mice or in response to calnexin shRNA reduced cellular NOX4 protein expression and reactive oxygen species formation. Our results suggest that endogenous NOX4 forms macromolecular complexes with calnexin, which are needed for the proper maturation, processing, and function of NOX4 in the endoplasmic reticulum.
Collapse
Affiliation(s)
- Kim-Kristin Prior
- From the Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Frankfurt am Main, 60590 Germany, the German Center for Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany
| | - Ilka Wittig
- the German Center for Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany the Functional Proteomics, SFB 815 Core Unit, Goethe-Universität, 60590 Frankfurt am Main, Germany, the Cluster of Excellence "Macromolecular Complexes," Goethe-Universität, 60590 Frankfurt am Main, Germany
| | - Matthias S Leisegang
- From the Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Frankfurt am Main, 60590 Germany, the German Center for Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany
| | - Jody Groenendyk
- the Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Norbert Weissmann
- the Excellence Cluster Cardio-Pulmonary System, Justus-Liebig-University Member of the German Center for Lung Research (DZL), 60590 Giessen, Germany
| | - Marek Michalak
- the Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Pidder Jansen-Dürr
- the Institute for Biomedical Ageing Research and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, 6020 Insbruk, Austria
| | - Ajay M Shah
- the King's College London British Heart Foundation Centre, Cardiovascular Division, London WC2R 2LS, United Kingdom, and
| | - Ralf P Brandes
- From the Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Frankfurt am Main, 60590 Germany, the German Center for Cardiovascular Research (DZHK), Partner site RheinMain, 60590 Frankfurt am Main, Germany
| |
Collapse
|
49
|
Chen F, Haigh S, Yu Y, Benson T, Wang Y, Li X, Dou H, Bagi Z, Verin AD, Stepp DW, Csanyi G, Chadli A, Weintraub NL, Smith SME, Fulton DJR. Nox5 stability and superoxide production is regulated by C-terminal binding of Hsp90 and CO-chaperones. Free Radic Biol Med 2015; 89:793-805. [PMID: 26456056 PMCID: PMC4751585 DOI: 10.1016/j.freeradbiomed.2015.09.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 08/15/2015] [Accepted: 09/03/2015] [Indexed: 10/22/2022]
Abstract
Heat shock protein 90 (Hsp90) is a molecular chaperone that orchestrates the folding and stability of proteins that regulate cellular signaling, proliferation and inflammation. We have previously shown that Hsp90 controls the production of reactive oxygen species by modulating the activity of Noxes1-3 and 5, but not Nox4. The goal of the current study was to define the regions on Nox5 that bind Hsp90 and determine how Hsp90 regulates enzyme activity. In isolated enzyme activity assays, we found that Hsp90 inhibitors selectively decrease superoxide, but not hydrogen peroxide, production. The addition of Hsp90 alone only modestly increases Nox5 enzyme activity but in combination with the co-chaperones, Hsp70, HOP, Hsp40, and p23 it robustly stimulated superoxide, but not hydrogen peroxide, production. Proximity ligation assays reveal that Nox5 and Hsp90 interact in intact cells. In cell lysates using a co-IP approach, Hsp90 binds to Nox5 but not Nox4, and the degree of binding can be influenced by calcium-dependent stimuli. Inhibition of Hsp90 induced the degradation of full length, catalytically inactive and a C-terminal fragment (aa398-719) of Nox5. In contrast, inhibition of Hsp90 did not affect the expression levels of N-terminal fragments (aa1-550) suggesting that Hsp90 binding maintains the stability of C-terminal regions. In Co-IP assays, Hsp90 was bound only to the C-terminal region of Nox5. Further refinement using deletion analysis revealed that the region between aa490-550 mediates Hsp90 binding. Converse mapping experiments show that the C-terminal region of Nox5 bound to the M domain of Hsp90 (aa310-529). In addition to Hsp90, Nox5 bound other components of the foldosome including co-chaperones Hsp70, HOP, p23 and Hsp40. Silencing of HOP, Hsp40 and p23 reduced Nox5-dependent superoxide. In contrast, increased expression of Hsp70 decreased Nox5 activity whereas a mutant of Hsp70 failed to do so. Inhibition of Hsp90 results in the loss of higher molecular weight complexes of Nox5 and decreased interaction between monomers. Collectively these results show that the C-terminal region of Nox5 binds to the M domain of Hsp90 and that the binding of Hsp90 and select co-chaperones facilitate oligomerization and the efficient production of superoxide.
Collapse
Affiliation(s)
- Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, 210029 China; Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA.
| | - Steven Haigh
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Yanfang Yu
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Tyler Benson
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Yusi Wang
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Xueyi Li
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Huijuan Dou
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Zsolt Bagi
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Alexander D Verin
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - David W Stepp
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Gabor Csanyi
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Ahmed Chadli
- Cancer Research Center, Molecular Chaperones Program, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Neal L Weintraub
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA
| | - Susan M E Smith
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw GA 30152, USA
| | - David J R Fulton
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia 30912, USA.
| |
Collapse
|
50
|
Craige SM, Kant S, Reif M, Chen K, Pei Y, Angoff R, Sugamura K, Fitzgibbons T, Keaney JF. Endothelial NADPH oxidase 4 protects ApoE-/- mice from atherosclerotic lesions. Free Radic Biol Med 2015; 89:1-7. [PMID: 26169727 PMCID: PMC4783146 DOI: 10.1016/j.freeradbiomed.2015.07.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 06/30/2015] [Accepted: 07/04/2015] [Indexed: 01/24/2023]
Abstract
Vascular reactive oxygen species (ROS) are known to be involved in atherosclerosis development and progression. NADPH oxidase 4 (Nox4) is a constitutively active ROS-producing enzyme that is highly expressed in the vascular endothelium. Nox4 is unique in its biology and has been implicated in vascular repair, however, the role of Nox4 in atherosclerosis is unknown. Therefore, to determine the effect of endothelial Nox4 on development of atherosclerosis, Apoe E-/- mice +/- endothelial Nox4 (ApoE-/- + EC Nox4) were fed a high cholesterol/high fat (Western) diet for 24 weeks. Significantly fewer atherosclerotic lesions were observed in the ApoE-/- + EC Nox4 mice as compared to the ApoE-/- littermates, which was most striking in the abdominal region of the aorta. In addition, markers of T cell populations were markedly different between the groups; T regulatory cell marker (FoxP3) was increased whereas T effector cell marker (T-bet) was decreased in aorta from ApoE-/- + EC Nox4 mice compared to ApoE-/- alone. We also observed decreased monokine induced by gamma interferon (MIG; CXCL9), a cytokine known to recruit and activate T cells, in plasma and tissue from ApoE-/- + EC Nox4 mice. To further investigate the link between endothelial Nox4 and MIG expression, we utilized cultured endothelial cells from our EC Nox4 transgenic mice and human cells with adenoviral overexpression of Nox4. In these cultured cells, upregulation of Nox4 attenuated endothelial cell MIG expression in response to interferon-gamma. Together these data suggest that endothelial Nox4 expression reduces MIG production and promotes a T cell distribution that favors repair over inflammation, leading to protection from atherosclerosis.
Collapse
Affiliation(s)
- Siobhan M Craige
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Shashi Kant
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Michaella Reif
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Kai Chen
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Yongmei Pei
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Rebecca Angoff
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Koichi Sugamura
- Department of Cardiovascular Medicine, Kumamoto University Hospital, Japan
| | - Timothy Fitzgibbons
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - John F Keaney
- Division of Cardiovascular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|