1
|
Xiao W, Lee LY, Loscalzo J. Metabolic Responses to Redox Stress in Vascular Cells. Antioxid Redox Signal 2024; 41:793-817. [PMID: 38985660 PMCID: PMC11876825 DOI: 10.1089/ars.2023.0476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/11/2023] [Indexed: 07/12/2024]
Abstract
Significance: Redox stress underlies numerous vascular disease mechanisms. Metabolic adaptability is essential for vascular cells to preserve energy and redox homeostasis. Recent Advances: Single-cell technologies and multiomic studies demonstrate significant metabolic heterogeneity among vascular cells in health and disease. Increasing evidence shows that reductive or oxidative stress can induce metabolic reprogramming of vascular cells. A recent example is intracellular L-2-hydroxyglutarate accumulation in response to hypoxic reductive stress, which attenuates the glucose flux through glycolysis and mitochondrial respiration in pulmonary vascular cells and provides protection against further reductive stress. Critical Issues: Regulation of cellular redox homeostasis is highly compartmentalized and complex. Vascular cells rely on multiple metabolic pathways, but the precise connectivity among these pathways and their regulatory mechanisms is only partially defined. There is also a critical need to understand better the cross-regulatory mechanisms between the redox system and metabolic pathways as perturbations in either systems or their cross talk can be detrimental. Future Directions: Future studies are needed to define further how multiple metabolic pathways are wired in vascular cells individually and as a network of closely intertwined processes given that a perturbation in one metabolic compartment often affects others. There also needs to be a comprehensive understanding of how different types of redox perturbations are sensed by and regulate different cellular metabolic pathways with specific attention to subcellular compartmentalization. Lastly, integration of dynamic changes occurring in multiple metabolic pathways and their cross talk with the redox system is an important goal in this multiomics era. Antioxid. Redox Signal. 41,793-817.
Collapse
Affiliation(s)
- Wusheng Xiao
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Toxicology, School of Public Health, Peking University, Beijing, China
| | - Laurel Y. Lee
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
3
|
Xuan Y, Yu C, Ni K, Congcong L, Lixin Q, Qingxian L. Protective effects of tanshinone IIA on Porphyromonas gingivalis-induced atherosclerosis via the downregulation of the NOX2/NOX4-ROS mediation of NF-κB signaling pathway. Microbes Infect 2023; 25:105177. [PMID: 37392987 DOI: 10.1016/j.micinf.2023.105177] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/07/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
Tanshinone IIA (TSA), an active component isolated from Danshen, possess high medicinal values against atherosclerosis by reducing vascular oxidative stress, inhibiting platelet aggregation, and protecting the endothelium from damage. The periodontal pathogen Porphyromonas gingivalis (P. gingivalis) has been proven to accelerate the development of atherosclerosis. We aim to determine the effects of TSA on P. gingivalis-induced atherosclerosis in ApoE-knockout (ApoE-/-) mice. After feeding with a high-lipid diet and infected with P. gingivalis three times per week for four weeks, TSA-treated (60 mg/kg/d) mice greatly inhibited atherosclerotic lesions both morphologically and biochemically and exhibited significantly reduction ROS, 8-OHdG, and ox-LDL levels in serum compared with P. gingivalis-infected mice. Additionally, TSA-treated mice were observed a marked reduction of ROS, 8-OHdG and ox-LDL in the serum, mRNA levels of COX-2, LOX-1, NOX2 and NOX4 in the aorta, as well as the levels of NOX2, NOX4, and NF-κB. These results suggest that TSA attenuates oxidative stress by decreasing NOX2 and NOX4 and downregulating NF-κB signaling pathway, which might be contributed to the amelioration of atherosclerosis.
Collapse
Affiliation(s)
- Yan Xuan
- Department of the Fourth Division, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Cai Yu
- Department of Periodontology, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Kang Ni
- Department of Periodontology, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Lou Congcong
- Department of Periodontology, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Qiu Lixin
- Department of the Fourth Division, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China.
| | - Luan Qingxian
- Department of Periodontology, Peking University, School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China.
| |
Collapse
|
4
|
Yu J, Qiu J, Zhang Z, Cui X, Guo W, Sheng M, Gao M, Wang D, Xu L, Ma X. Redox Biology in Adipose Tissue Physiology and Obesity. Adv Biol (Weinh) 2023; 7:e2200234. [PMID: 36658733 DOI: 10.1002/adbi.202200234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/24/2022] [Indexed: 01/21/2023]
Abstract
Reactive oxygen species (ROS), a by-product of mitochondrial oxidative phosphorylation and cellular metabolism, is vital for cellular survival, proliferation, damage, and senescence. In recent years, studies have shown that ROS levels and redox status in adipose tissue are strongly associated with obesity and metabolic diseases. Although it was previously considered that excessive production of ROS and impairment of antioxidant capability leads to oxidative stress and potentially contributes to increased adiposity, it has become increasingly evident that an adequate amount of ROS is vital for adipocyte differentiation and thermogenesis. In this review, by providing a systematic overview of the recent understanding of the key factors of redox systems, endogenous mechanisms for redox homeostasis, advanced techniques for dynamic redox monitoring, as well as exogenous stimuli for redox production in adipose tissues and obesity, the importance of redox biology in metabolic health is emphasized.
Collapse
Affiliation(s)
- Jian Yu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai, 201499, P. R. China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Jin Qiu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Zhe Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Xiangdi Cui
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Wenxiu Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Maozheng Sheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Mingyuan Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
| | - Xinran Ma
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai, 201499, P. R. China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, P. R. China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, P. R. China
| |
Collapse
|
5
|
Demircan MB, Schnoeder TM, Mgbecheta PC, Schröder K, Böhmer FD, Heidel FH. Context-specific effects of NOX4 inactivation in acute myeloid leukemia (AML). J Cancer Res Clin Oncol 2022; 148:1983-1990. [PMID: 35348887 PMCID: PMC9293823 DOI: 10.1007/s00432-022-03986-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE Oxidative stress has been linked to initiation and progression of cancer and recent studies have indicated a potential translational role regarding modulation of ROS in various cancers, including acute myeloid leukemia (AML). Detailed understanding of the complex machinery regulating ROS including its producer elements in cancer is required to define potential translational therapeutic use. Based on previous studies in acute myeloid leukemia (AML) models, we considered NADPH oxidase (NOX) family members, specifically NOX4 as a potential target in AML. METHODS Pharmacologic inhibition and genetic inactivation of NOX4 in murine and human models of AML were used to understand its functional role. For genetic inactivation, CRISPR-Cas9 technology was used in human AML cell lines in vitro and genetically engineered knockout mice for Nox4 were used for deletion of Nox4 in hematopoietic cells via Mx1-Cre recombinase activation. RESULTS Pharmacologic NOX inhibitors and CRISPR-Cas9-mediated inactivation of NOX4 and p22-phox (an essential NOX component) decreased proliferative capacity and cell competition in FLT3-ITD-positive human AML cells. In contrast, conditional deletion of Nox4 enhanced the myeloproliferative phenotype of an FLT3-ITD induced knock-in mouse model. Finally, Nox4 inactivation in normal hematopoietic stem and progenitor cells (HSPCs) caused a minor reduction in HSC numbers and reconstitution capacity. CONCLUSION The role of NOX4 in myeloid malignancies appears highly context-dependent and its inactivation results in either enhancing or inhibitory effects. Therefore, targeting NOX4 in FLT3-ITD positive myeloid malignancies requires additional pre-clinical assessment.
Collapse
Affiliation(s)
- Muhammed Burak Demircan
- Innere Medizin II, Hämatologie und Onkologie, Jena University Hospital, Jena, Germany
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Tina M Schnoeder
- Innere Medizin C, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Peter C Mgbecheta
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Frank-D Böhmer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Florian H Heidel
- Innere Medizin II, Hämatologie und Onkologie, Jena University Hospital, Jena, Germany.
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany.
- Innere Medizin C, Universitätsmedizin Greifswald, Greifswald, Germany.
| |
Collapse
|
6
|
Shannon N, Gravelle R, Cunniff B. Mitochondrial trafficking and redox/phosphorylation signaling supporting cell migration phenotypes. Front Mol Biosci 2022; 9:925755. [PMID: 35936783 PMCID: PMC9355248 DOI: 10.3389/fmolb.2022.925755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Regulation of cell signaling cascades is critical in making sure the response is activated spatially and for a desired duration. Cell signaling cascades are spatially and temporally controlled through local protein phosphorylation events which are determined by the activation of specific kinases and/or inactivation of phosphatases to elicit a complete and thorough response. For example, A-kinase-anchoring proteins (AKAPs) contribute to the local regulated activity protein kinase A (PKA). The activity of kinases and phosphatases can also be regulated through redox-dependent cysteine modifications that mediate the activity of these proteins. A primary example of this is the activation of the epidermal growth factor receptor (EGFR) and the inactivation of the phosphatase and tensin homologue (PTEN) phosphatase by reactive oxygen species (ROS). Therefore, the local redox environment must play a critical role in the timing and magnitude of these events. Mitochondria are a primary source of ROS and energy (ATP) that contributes to redox-dependent signaling and ATP-dependent phosphorylation events, respectively. The strategic positioning of mitochondria within cells contributes to intracellular gradients of ROS and ATP, which have been shown to correlate with changes to protein redox and phosphorylation status driving downstream cellular processes. In this review, we will discuss the relationship between subcellular mitochondrial positioning and intracellular ROS and ATP gradients that support dynamic oxidation and phosphorylation signaling and resulting cellular effects, specifically associated with cell migration signaling.
Collapse
Affiliation(s)
- Nathaniel Shannon
- Department of Pathology and Laboratory Medicine, Redox Biology Program, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Randi Gravelle
- Department of Pathology and Laboratory Medicine, Redox Biology Program, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, Redox Biology Program, University of Vermont Larner College of Medicine, Burlington, VT, United States
- University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT, United States
| |
Collapse
|
7
|
Guo CL. Self-Sustained Regulation or Self-Perpetuating Dysregulation: ROS-dependent HIF-YAP-Notch Signaling as a Double-Edged Sword on Stem Cell Physiology and Tumorigenesis. Front Cell Dev Biol 2022; 10:862791. [PMID: 35774228 PMCID: PMC9237464 DOI: 10.3389/fcell.2022.862791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/29/2022] [Indexed: 12/19/2022] Open
Abstract
Organ development, homeostasis, and repair often rely on bidirectional, self-organized cell-niche interactions, through which cells select cell fate, such as stem cell self-renewal and differentiation. The niche contains multiplexed chemical and mechanical factors. How cells interpret niche structural information such as the 3D topology of organs and integrate with multiplexed mechano-chemical signals is an open and active research field. Among all the niche factors, reactive oxygen species (ROS) have recently gained growing interest. Once considered harmful, ROS are now recognized as an important niche factor in the regulation of tissue mechanics and topology through, for example, the HIF-YAP-Notch signaling pathways. These pathways are not only involved in the regulation of stem cell physiology but also associated with inflammation, neurological disorder, aging, tumorigenesis, and the regulation of the immune checkpoint molecule PD-L1. Positive feedback circuits have been identified in the interplay of ROS and HIF-YAP-Notch signaling, leading to the possibility that under aberrant conditions, self-organized, ROS-dependent physiological regulations can be switched to self-perpetuating dysregulation, making ROS a double-edged sword at the interface of stem cell physiology and tumorigenesis. In this review, we discuss the recent findings on how ROS and tissue mechanics affect YAP-HIF-Notch-PD-L1 signaling, hoping that the knowledge can be used to design strategies for stem cell-based and ROS-targeting therapy and tissue engineering.
Collapse
Affiliation(s)
- Chin-Lin Guo
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
8
|
Hendricks KS, To EE, Luong R, Liong F, Erlich JR, Shah AM, Liong S, O’Leary JJ, Brooks DA, Vlahos R, Selemidis S. Endothelial NOX4 Oxidase Negatively Regulates Inflammation and Improves Morbidity During Influenza A Virus Lung Infection in Mice. Front Cell Infect Microbiol 2022; 12:883448. [PMID: 35601109 PMCID: PMC9115386 DOI: 10.3389/fcimb.2022.883448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/07/2022] [Indexed: 12/02/2022] Open
Abstract
Endosomal NOX2 oxidase-dependent ROS production promotes influenza pathogenicity, but the role of NOX4 oxidase, which is highly expressed in the lung endothelium, is largely unknown. The aim of this study was to determine if endothelial NOX4 expression can influence viral pathology in vivo, using a mouse model of influenza infection. WT and transgenic endothelial NOX4 overexpressing mice (NOX4 TG) were infected intranasally with the Hong Kong H3N2 X-31 influenza A virus (104 PFU; HK x-31) or PBS control. Mice were culled at either 3 or 7 days post-infection to analyse: airway inflammation by bronchoalveolar lavage fluid (BALF) cell counts; NOX4, as well as inflammatory cytokine and chemokine gene expression by QPCR; and ROS production by an L-012-enhanced chemiluminescence assay. Influenza A virus infection of WT mice resulted in a significant reduction in lung NOX4 mRNA at day 3, which persisted until day 7, when compared to uninfected mice. Influenza A virus infection of NOX4 TG mice resulted in significantly less weight loss than that of WT mice at 3-days post infection. Viral titres were decreased in infected NOX4 TG mice compared to the infected WT mice, at both 3- and 7-days post infection and there was significantly less lung alveolitis, peri-bronchial inflammation and neutrophil infiltration. The oxidative burst from BALF inflammatory cells extracted from infected NOX4 TG mice was significantly less than that in the WT mice. Expression of macrophage and neutrophil chemoattractants CXCL10, CCL3, CXCL1 and CXCL2 in the lung tissue were significantly lower in NOX4 TG mice compared to the WT mice at 3-days post infection. We conclude that endothelial NOX4 oxidase is protective against influenza morbidity and is a potential target for limiting influenza A virus-induced lung inflammation.
Collapse
Affiliation(s)
- Keshia S. Hendricks
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Eunice E. To
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Raymond Luong
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Felicia Liong
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Jonathan R. Erlich
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Ajay M. Shah
- King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Stella Liong
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - John J. O’Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, St. James’s Hospital Dublin, Dublin, Ireland
| | - Doug A. Brooks
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| |
Collapse
|
9
|
Liu J, Wang L, Ge L, Sun W, Song Z, Lu X, Jin C, Wu S, Yang J. Lanthanum decreased VAPB-PTPP51, BAP31-FIS1, and MFN2-MFN1 expression of mitochondria-associated membranes and induced abnormal autophagy in rat hippocampus. Food Chem Toxicol 2022; 161:112831. [DOI: 10.1016/j.fct.2022.112831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/03/2022] [Accepted: 01/20/2022] [Indexed: 12/21/2022]
|
10
|
Characterization of a murine model of endothelial dysfunction induced by chronic intraperitoneal administration of angiotensin II. Sci Rep 2021; 11:21193. [PMID: 34707201 PMCID: PMC8551243 DOI: 10.1038/s41598-021-00676-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/14/2021] [Indexed: 02/05/2023] Open
Abstract
Endothelial dysfunction (ED) is a key factor for the development of cardiovascular diseases. Due to its chronic, life-threatening nature, ED only can be studied experimentally in animal models. Therefore, this work was aimed to characterize a murine model of ED induced by a daily intraperitoneal administration of angiotensin II (AGII) for 10 weeks. Oxidative stress, inflammation, vascular remodeling, hypertension, and damage to various target organs were evaluated in treated animals. The results indicated that a chronic intraperitoneal administration of AGII increases the production of systemic soluble VCAM, ROS and ICAM-1 expression, and the production of TNFα, IL1β, IL17A, IL4, TGFβ, and IL10 in the kidney, as well as blood pressure levels; it also promotes vascular remodeling and induces non-alcoholic fatty liver disease, glomerulosclerosis, and proliferative retinopathy. Therefore, the model herein proposed can be a representative model for ED; additionally, it is easy to implement, safe, rapid, and inexpensive.
Collapse
|
11
|
Bakr MH, Radwan E, Shaltout AS, Farrag AA, Mahmoud AR, Abd-Elhamid TH, Ali M. Chronic exposure to tramadol induces cardiac inflammation and endothelial dysfunction in mice. Sci Rep 2021; 11:18772. [PMID: 34548593 PMCID: PMC8455605 DOI: 10.1038/s41598-021-98206-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/06/2021] [Indexed: 11/21/2022] Open
Abstract
Tramadol is an opioid extensively used to treat moderate to severe pain; however, prolonged therapy is associated with several tissues damage. Chronic use of tramadol was linked to increased hospitalizations due to cardiovascular complications. Limited literature has described the effects of tramadol on the cardiovascular system, so we sought to investigate these actions and elucidate the underlying mechanisms. Mice received tramadol hydrochloride (40 mg/kg body weight) orally for 4 successive weeks. Oxidative stress, inflammation, and cardiac toxicity were assessed. In addition, eNOS expression was evaluated. Our results demonstrated marked histopathological alteration in heart and aortic tissues after exposure to tramadol. Tramadol upregulated the expression of oxidative stress and inflammatory markers in mice heart and aorta, whereas downregulated eNOS expression. Tramadol caused cardiac damage shown by the increase in LDH, Troponin I, and CK-MB activities in serum samples. Overall, these results highlight the risks of tramadol on the cardiovascular system.
Collapse
Affiliation(s)
- Marwa H Bakr
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt.
| | - Eman Radwan
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt.,Department of Biochemistry, Sphinx University, Assiut, Egypt
| | - Asmaa S Shaltout
- Department of Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Alshaimaa A Farrag
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt.,Department of Anatomy, College of Medicine, Bisha University, Bisha, Kingdom of Saudi Arabia
| | - Amany Refaat Mahmoud
- Department of Human Anatomy and Embryology, Faculty of Medicine, Assiut University, Assiut, Egypt.,Department of Basic Medical Sciences, Unaizah College of Medicine and Medical Sciences, Qassim University, Unaizah, Kingdom of Saudi Arabia
| | - Tarek Hamdy Abd-Elhamid
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Maha Ali
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
12
|
Viswambharan H, Yuldasheva NY, Imrie H, Bridge K, Haywood NJ, Skromna A, Hemmings KE, Clark ER, Gatenby VK, Cordell P, Simmons KJ, Makava N, Abudushalamu Y, Endesh N, Brown J, Walker AMN, Futers ST, Porter KE, Cubbon RM, Naseem K, Shah AM, Beech DJ, Wheatcroft SB, Kearney MT, Sukumar P. Novel Paracrine Action of Endothelium Enhances Glucose Uptake in Muscle and Fat. Circ Res 2021; 129:720-734. [PMID: 34420367 PMCID: PMC8448413 DOI: 10.1161/circresaha.121.319517] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hema Viswambharan
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Nadira Y Yuldasheva
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Helen Imrie
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Katherine Bridge
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Natalie J Haywood
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Anna Skromna
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Karen E Hemmings
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Emily R Clark
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - V Kate Gatenby
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Paul Cordell
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Katie J Simmons
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Natallia Makava
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Yilizila Abudushalamu
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Naima Endesh
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Jane Brown
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Andrew M N Walker
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Simon T Futers
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Karen E Porter
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Richard M Cubbon
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Khalid Naseem
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Ajay M Shah
- British Heart Foundation Centre of Research Excellence, King's College London (A.M.S.)
| | - David J Beech
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Stephen B Wheatcroft
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| | - Piruthivi Sukumar
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., H.I., K.B., N.J.H., A.S., K.E.H., E.R.C., V.K.G., P.C., K.J.S., N.M., Y.A., N.E., J.B., A.M.N.W., S.T.F., K.E.P., R.M.C., K.N., D.J.B., S.B.W., M.T.K., P.S.)
| |
Collapse
|
13
|
Martins-Marques T, Rodriguez-Sinovas A, Girao H. Cellular crosstalk in cardioprotection: Where and when do reactive oxygen species play a role? Free Radic Biol Med 2021; 169:397-409. [PMID: 33892116 DOI: 10.1016/j.freeradbiomed.2021.03.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/14/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022]
Abstract
A well-balanced intercellular communication between the different cells within the heart is vital for the maintenance of cardiac homeostasis and function. Despite remarkable advances on disease management and treatment, acute myocardial infarction remains the major cause of morbidity and mortality worldwide. Gold standard reperfusion strategies, namely primary percutaneous coronary intervention, are crucial to preserve heart function. However, reestablishment of blood flow and oxygen levels to the infarcted area are also associated with an accumulation of reactive oxygen species (ROS), leading to oxidative damage and cardiomyocyte death, a phenomenon termed myocardial reperfusion injury. In addition, ROS signaling has been demonstrated to regulate multiple biological pathways, including cell differentiation and intercellular communication. Given the importance of cell-cell crosstalk in the coordinated response after cell injury, in this review, we will discuss the impact of ROS in the different forms of inter- and intracellular communication, as well as the role of gap junctions, tunneling nanotubes and extracellular vesicles in the propagation of oxidative damage in cardiac diseases, particularly in the context of ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Tania Martins-Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Antonio Rodriguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall D'Hebron Institut de Recerca (VHIR), Vall D'Hebron Hospital Universitari, Vall D'Hebron Barcelona Hospital Campus, Passeig Vall D'Hebron, 119-129, 08035, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Henrique Girao
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
14
|
Thallas-Bonke V, Tan SM, Lindblom RS, Snelson M, Granata C, Jha JC, Sourris KC, Laskowski A, Watson A, Tauc M, Rubera I, Zheng G, Shah AM, Harris DCH, Elbatreek MH, Kantharidis P, Cooper ME, Jandeleit-Dahm K, Coughlan MT. Targeted deletion of nicotinamide adenine dinucleotide phosphate oxidase 4 from proximal tubules is dispensable for diabetic kidney disease development. Nephrol Dial Transplant 2021; 36:988-997. [PMID: 33367789 DOI: 10.1093/ndt/gfaa376] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The nicotinamide adenine dinucleotide phosphate oxidase isoform 4 (Nox4) mediates reactive oxygen species (ROS) production and renal fibrosis in diabetic kidney disease (DKD) at the level of the podocyte. However, the mitochondrial localization of Nox4 and its role as a mitochondrial bioenergetic sensor has recently been reported. Whether Nox4 drives pathology in DKD within the proximal tubular compartment, which is densely packed with mitochondria, is not yet known. METHODS We generated a proximal tubular-specific Nox4 knockout mouse model by breeding Nox4flox/flox mice with mice expressing Cre recombinase under the control of the sodium-glucose cotransporter-2 promoter. Subsets of Nox4ptKO mice and their Nox4flox/flox littermates were injected with streptozotocin (STZ) to induce diabetes. Mice were followed for 20 weeks and renal injury was assessed. RESULTS Genetic ablation of proximal tubular Nox4 (Nox4ptKO) resulted in no change in renal function and histology. Nox4ptKO mice and Nox4flox/flox littermates injected with STZ exhibited the hallmarks of DKD, including hyperfiltration, albuminuria, renal fibrosis and glomerulosclerosis. Surprisingly, diabetes-induced renal injury was not improved in Nox4ptKO STZ mice compared with Nox4flox/flox STZ mice. Although diabetes conferred ROS overproduction and increased the mitochondrial oxygen consumption rate, proximal tubular deletion of Nox4 did not normalize oxidative stress or mitochondrial bioenergetics. CONCLUSIONS Taken together, these results demonstrate that genetic deletion of Nox4 from the proximal tubules does not influence DKD development, indicating that Nox4 localization within this highly energetic compartment is dispensable for chronic kidney disease pathogenesis in the setting of diabetes.
Collapse
Affiliation(s)
| | - Sih Min Tan
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Runa S Lindblom
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Matthew Snelson
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Cesare Granata
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Jay Chandra Jha
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Karly C Sourris
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Adrienne Laskowski
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Anna Watson
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Michel Tauc
- Laboratoire de Physiomédecine Moléculaire, LP2M, UMR-CNRS 7370, Université Côte d'Azur, Nice, France
| | - Isabelle Rubera
- Laboratoire de Physiomédecine Moléculaire, LP2M, UMR-CNRS 7370, Université Côte d'Azur, Nice, France
| | - Guoping Zheng
- Centre for Transplantation and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine and Sciences, London, UK
| | - David C H Harris
- Centre for Transplantation and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Mahmoud H Elbatreek
- Department of Pharmacology and Personalised Medicine, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Phillip Kantharidis
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Mark E Cooper
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Karin Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
- German Diabetes Centre, Leibniz Centre for Diabetes Research, Heinrich Heine University, Duesseldorf, Germany
| | - Melinda T Coughlan
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Diabetes, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| |
Collapse
|
15
|
Mustapha S, Mohammed M, Azemi AK, Yunusa I, Shehu A, Mustapha L, Wada Y, Ahmad MH, Ahmad WANW, Rasool AHG, Mokhtar SS. Potential Roles of Endoplasmic Reticulum Stress and Cellular Proteins Implicated in Diabesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8830880. [PMID: 33995826 PMCID: PMC8099518 DOI: 10.1155/2021/8830880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/28/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
The role of the endoplasmic reticulum (ER) has evolved from protein synthesis, processing, and other secretory pathways to forming a foundation for lipid biosynthesis and other metabolic functions. Maintaining ER homeostasis is essential for normal cellular function and survival. An imbalance in the ER implied stressful conditions such as metabolic distress, which activates a protective process called unfolded protein response (UPR). This response is activated through some canonical branches of ER stress, i.e., the protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1α (IRE1α), and activating transcription factor 6 (ATF6). Therefore, chronic hyperglycemia, hyperinsulinemia, increased proinflammatory cytokines, and free fatty acids (FFAs) found in diabesity (a pathophysiological link between obesity and diabetes) could lead to ER stress. However, limited data exist regarding ER stress and its association with diabesity, particularly the implicated proteins and molecular mechanisms. Thus, this review highlights the role of ER stress in relation to some proteins involved in diabesity pathogenesis and provides insight into possible pathways that could serve as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mustapha Mohammed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Pulau Pinang, Malaysia
- Department of Clinical Pharmacy and Pharmacy Practice, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Ismaeel Yunusa
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina, College of Pharmacy, Columbia, SC, USA
| | - Aishatu Shehu
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Lukman Mustapha
- Department of Pharmaceutical and Medicinal Chemistry, Kaduna State University, Kaduna, Nigeria
| | - Yusuf Wada
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Department of Zoology, Ahmadu Bello University Zaria, Kaduna, Nigeria
| | - Mubarak Hussaini Ahmad
- Department of Pharmacology and Therapeutics, Ahmadu Bello University Zaria, Kaduna, Nigeria
- School of Pharmacy Technician, Aminu Dabo College of Health Sciences and Technology, Kano, Nigeria
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
16
|
Wouk J, Dekker RFH, Queiroz EAIF, Barbosa-Dekker AM. β-Glucans as a panacea for a healthy heart? Their roles in preventing and treating cardiovascular diseases. Int J Biol Macromol 2021; 177:176-203. [PMID: 33609583 DOI: 10.1016/j.ijbiomac.2021.02.087] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Factors increasing the risks for CVD development are related to obesity, diabetes, high blood cholesterol, high blood pressure and lifestyle. CVD risk factors may be treated with appropriate drugs, but prolonged can use cause undesirable side-effects. Among the natural products used in complementary and alternative medicines, are the β-ᴅ-glucans; biopolymers found in foods (cereals, mushrooms), and can easily be produced by microbial fermentation. Independent of source, β-glucans of the mixed-linked types [(1 → 3)(1 → 6)-β-ᴅ-glucans - fungal, and (1 → 3)(1 → 4)-β-ᴅ-glucans - cereal] have widely been studied because of their biological activities, and have demonstrated cardiovascular protective effects. In this review, we discuss the roles of β-ᴅ-glucans in various pathophysiological conditions that lead to CVDs including obesity, dyslipidemia, hyperglycemia, oxidative stress, hypertension, atherosclerosis and stroke. The β-glucans from all of the sources cited demonstrated potential hypoglycemic, hypocholesterolemic and anti-obesogenicity activities, reduced hypertension and ameliorated the atherosclerosis condition. More recently, β-glucans are recognized as possessing prebiotic properties that modulate the gut microbiome and impact on the health benefits including cardiovascular. Overall, all the studies investigated unequivocally demonstrated the dietary benefits of consuming β-glucans regardless of source, thus constituting a promising panaceutical approach to reduce CVD risk factors.
Collapse
Affiliation(s)
- Jéssica Wouk
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Estadual do Centro-Oeste, Campus CEDETEG, CEP: 85040-167, Guarapuava, Paraná, Brazil
| | - Robert F H Dekker
- Universidade Tecnológica Federal do Paraná, Programa de Pós-Graduação em Engenharia Ambiental, Câmpus Londrina, CEP: 86036-370 Londrina, Paraná, Brazil; Beta-Glucan Produtos Farmoquímicos - EIRELI, Avenida João Miguel Caram 731, Lote 24(A), Bloco Zircônia, Universidade Tecnológica Federal do Paraná, CEP: 86036-700 Londrina, Paraná, Brazil.
| | - Eveline A I F Queiroz
- Núcleo de Pesquisa e Apoio Didático em Saúde, Instituto de Ciências da Saúde, Câmpus Universitário de Sinop, Universidade Federal de Mato Grosso, CEP: 78.557-267 Sinop, Mato Grosso, Brazil
| | - Aneli M Barbosa-Dekker
- Beta-Glucan Produtos Farmoquímicos - EIRELI, Avenida João Miguel Caram 731, Lote 24(A), Bloco Zircônia, Universidade Tecnológica Federal do Paraná, CEP: 86036-700 Londrina, Paraná, Brazil
| |
Collapse
|
17
|
Maietta V, Reyes-García J, Yadav VR, Zheng YM, Peng X, Wang YX. Cellular and Molecular Processes in Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:21-38. [PMID: 34019261 DOI: 10.1007/978-3-030-68748-9_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pulmonary hypertension (PH) is a progressive lung disease characterized by persistent pulmonary vasoconstriction. Another well-recognized characteristic of PH is the muscularization of peripheral pulmonary arteries. This pulmonary vasoremodeling manifests in medial hypertrophy/hyperplasia of smooth muscle cells (SMCs) with possible neointimal formation. The underlying molecular processes for these two major vascular responses remain not fully understood. On the other hand, a series of very recent studies have shown that the increased reactive oxygen species (ROS) seems to be an important player in mediating pulmonary vasoconstriction and vasoremodeling, thereby leading to PH. Mitochondria are a primary site for ROS production in pulmonary artery (PA) SMCs, which subsequently activate NADPH oxidase to induce further ROS generation, i.e., ROS-induced ROS generation. ROS control the activity of multiple ion channels to induce intracellular Ca2+ release and extracellular Ca2+ influx (ROS-induced Ca2+ release and influx) to cause PH. ROS and Ca2+ signaling may synergistically trigger an inflammatory cascade to implicate in PH. Accordingly, this paper explores the important roles of ROS, Ca2+, and inflammatory signaling in the development of PH, including their reciprocal interactions, key molecules, and possible therapeutic targets.
Collapse
Affiliation(s)
- Vic Maietta
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Jorge Reyes-García
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA.,Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Vishal R Yadav
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Yun-Min Zheng
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA.
| | - Xu Peng
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, TX, USA.
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
18
|
Schnelle M, Sawyer I, Anilkumar N, Mohamed BA, Richards DA, Toischer K, Zhang M, Catibog N, Sawyer G, Mongue-Din H, Schröder K, Hasenfuss G, Shah AM. NADPH oxidase-4 promotes eccentric cardiac hypertrophy in response to volume overload. Cardiovasc Res 2021; 117:178-187. [PMID: 31821410 PMCID: PMC7797217 DOI: 10.1093/cvr/cvz331] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/13/2019] [Accepted: 12/07/2019] [Indexed: 12/15/2022] Open
Abstract
AIMS Chronic pressure or volume overload induce concentric vs. eccentric left ventricular (LV) remodelling, respectively. Previous studies suggest that distinct signalling pathways are involved in these responses. NADPH oxidase-4 (Nox4) is a reactive oxygen species-generating enzyme that can limit detrimental cardiac remodelling in response to pressure overload. This study aimed to assess its role in volume overload-induced remodelling. METHODS AND RESULTS We compared the responses to creation of an aortocaval fistula (Shunt) to induce volume overload in Nox4-null mice (Nox4-/-) vs. wild-type (WT) littermates. Induction of Shunt resulted in a significant increase in cardiac Nox4 mRNA and protein levels in WT mice as compared to Sham controls. Nox4-/- mice developed less eccentric LV remodelling than WT mice (echocardiographic relative wall thickness: 0.30 vs. 0.27, P < 0.05), with less LV hypertrophy at organ level (increase in LV weight/tibia length ratio of 25% vs. 43%, P < 0.01) and cellular level (cardiomyocyte cross-sectional area: 323 µm2 vs. 379 μm2, P < 0.01). LV ejection fraction, foetal gene expression, interstitial fibrosis, myocardial capillary density, and levels of myocyte apoptosis after Shunt were similar in the two genotypes. Myocardial phospho-Akt levels were increased after induction of Shunt in WT mice, whereas levels decreased in Nox4-/- mice (+29% vs. -21%, P < 0.05), associated with a higher level of phosphorylation of the S6 ribosomal protein (S6) and the eIF4E-binding protein 1 (4E-BP1) in WT compared to Nox4-/- mice. We identified that Akt activation in cardiac cells is augmented by Nox4 via a Src kinase-dependent inactivation of protein phosphatase 2A. CONCLUSION Endogenous Nox4 is required for the full development of eccentric cardiac hypertrophy and remodelling during chronic volume overload. Nox4-dependent activation of Akt and its downstream targets S6 and 4E-BP1 may be involved in this effect.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Apoptosis
- Arteriovenous Shunt, Surgical
- Cell Cycle Proteins/metabolism
- Cell Line
- Disease Models, Animal
- Fibrosis
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- NADPH Oxidase 2/genetics
- NADPH Oxidase 2/metabolism
- NADPH Oxidase 4/genetics
- NADPH Oxidase 4/metabolism
- Phosphorylation
- Protein Phosphatase 2/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Ribosomal Protein S6/metabolism
- Signal Transduction
- Ventricular Function, Left
- Ventricular Remodeling
- src-Family Kinases/metabolism
- Mice
Collapse
Affiliation(s)
- Moritz Schnelle
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- Institute for Clinical Chemistry, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Iain Sawyer
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Narayana Anilkumar
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Daniel A Richards
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Min Zhang
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Norman Catibog
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Greta Sawyer
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Héloïse Mongue-Din
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Ajay M Shah
- King’s College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
19
|
Graceffa V. Therapeutic Potential of Reactive Oxygen Species: State of the Art and Recent Advances. SLAS Technol 2020; 26:140-158. [PMID: 33345675 DOI: 10.1177/2472630320977450] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In the last decade, several studies have proven that when at low concentration reactive oxygen species (ROS) show an adaptive beneficial effect and posited the idea that they can be utilized as inexpensive and convenient inducers of tissue regeneration. On the other hand, the recent discovery that cancer cells are more sensitive to oxidative damage paved the way for their use in the selective killing of tumor cells, and sensors to monitor ROS production during cancer treatment are under extensive investigation. Nevertheless, although ROS-activated signaling pathways are well established, less is known about the mechanisms underlying the switch from an anabolic to a cytotoxic response. Furthermore, a high variability in biological response is observed between different modalities of administration, cell types, donor ages, eventual concomitant diseases, and external microenvironment. On the other hand, available preclinical studies are scarce, whereas the quest for the most suitable systems for in vivo delivery is still elusive. Furthermore, new strategies to control the temporal pattern of ROS release need to be developed, if considering their tumorigenic potential. This review initially discusses ROS mechanisms of action and their potential application in stem cell biology, tissue engineering, and cancer therapy. It then outlines the state of art of ROS-based drugs and identifies challenges faced in translating ROS research into clinical practice.
Collapse
Affiliation(s)
- Valeria Graceffa
- Cellular Health and Toxicology Research Group (CHAT), Institute of Technology Sligo, Bellanode, Sligo, Ireland.,Department of Life Sciences, Institute of Technology Sligo, Bellanode, Sligo, Ireland
| |
Collapse
|
20
|
Konst RE, Guzik TJ, Kaski JC, Maas AHEM, Elias-Smale SE. The pathogenic role of coronary microvascular dysfunction in the setting of other cardiac or systemic conditions. Cardiovasc Res 2020; 116:817-828. [PMID: 31977015 PMCID: PMC7526753 DOI: 10.1093/cvr/cvaa009] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/09/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Coronary microvascular dysfunction (CMD) plays a pathogenic role in cardiac and systemic conditions other than microvascular angina. In this review, we provide an overview of the pathogenic role of CMD in the setting of diabetes mellitus, obesity, hypertensive pregnancy disorders, chronic inflammatory and autoimmune rheumatic disorders, chronic kidney disease, hypertrophic cardiomyopathy, and aortic valve stenosis. In these various conditions, CMD results from different structural, functional, and/or dynamic alterations in the coronary microcirculation associated with the primary disease process. CMD is often detectable very early in the course of the primary disease, before clinical symptoms or signs of myocardial ischaemia are present, and it portrays an increased risk for cardiovascular events.
Collapse
Affiliation(s)
- Regina E Konst
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow, UK
| | - Juan-Carlos Kaski
- The Queen Elizabeth Hospital Discipline of Medicine, University of Adelaide, Central Adelaide Local Health Network, Coronary Vasomotion Disorders International Study Group (COVADIS), Adelaide, Australia.,Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Angela H E M Maas
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Suzette E Elias-Smale
- Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
21
|
Beretta M, Santos CXC, Molenaar C, Hafstad AD, Miller CCJ, Revazian A, Betteridge K, Schröder K, Streckfuß‐Bömeke K, Doroshow JH, Fleck RA, Su T, Belousov VV, Parsons M, Shah AM. Nox4 regulates InsP 3 receptor-dependent Ca 2+ release into mitochondria to promote cell survival. EMBO J 2020; 39:e103530. [PMID: 33001475 PMCID: PMC7527947 DOI: 10.15252/embj.2019103530] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 06/22/2020] [Accepted: 07/01/2020] [Indexed: 12/24/2022] Open
Abstract
Cells subjected to environmental stresses undergo regulated cell death (RCD) when homeostatic programs fail to maintain viability. A major mechanism of RCD is the excessive calcium loading of mitochondria and consequent triggering of the mitochondrial permeability transition (mPT), which is especially important in post-mitotic cells such as cardiomyocytes and neurons. Here, we show that stress-induced upregulation of the ROS-generating protein Nox4 at the ER-mitochondria contact sites (MAMs) is a pro-survival mechanism that inhibits calcium transfer through InsP3 receptors (InsP3 R). Nox4 mediates redox signaling at the MAM of stressed cells to augment Akt-dependent phosphorylation of InsP3 R, thereby inhibiting calcium flux and mPT-dependent necrosis. In hearts subjected to ischemia-reperfusion, Nox4 limits infarct size through this mechanism. These results uncover a hitherto unrecognized stress pathway, whereby a ROS-generating protein mediates pro-survival effects through spatially confined signaling at the MAM to regulate ER to mitochondria calcium flux and triggering of the mPT.
Collapse
Affiliation(s)
- Matteo Beretta
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| | - Celio XC Santos
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| | - Chris Molenaar
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| | - Anne D Hafstad
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
- Cardiovascular Research GroupDepartment of Medical BiologyUIT‐The Arctic University of NorwayTromsøNorway
| | - Chris CJ Miller
- Department of Basic and Clinical NeuroscienceInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Aram Revazian
- Institute for Cardiovascular PhysiologyGeorg August University GöttingenGöttingenGermany
| | - Kai Betteridge
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| | - Katrin Schröder
- Institute for Cardiovascular PhysiologyGoethe‐University FrankfurtFrankfurt am MainGermany
| | | | - James H Doroshow
- Division of Cancer Treatment and DiagnosisNational Cancer InstituteNIHBethesdaMDUSA
| | - Roland A Fleck
- Centre for Ultrastructural ImagingKing's College LondonLondonUK
| | - Tsung‐Ping Su
- Cellular Pathobiology SectionNational Institute on Drug Abuse, NIHBaltimoreMDUSA
| | - Vsevolod V Belousov
- Institute for Cardiovascular PhysiologyGeorg August University GöttingenGöttingenGermany
- Shemyakin‐Ovchinnikov Institute of Bioorganic ChemistryMoscowRussia
- Pirogov Russian National Research Medical UniversityMoscowRussia
| | - Maddy Parsons
- King's College London British Heart Foundation CentreRandall Centre of Cell and Molecular BiophysicsLondonUK
| | - Ajay M Shah
- School of Cardiovascular Medicine & SciencesKing's College London British Heart Foundation CentreLondonUK
| |
Collapse
|
22
|
Takamura T. Hepatokine Selenoprotein P-Mediated Reductive Stress Causes Resistance to Intracellular Signal Transduction. Antioxid Redox Signal 2020; 33:517-524. [PMID: 32295394 PMCID: PMC7409583 DOI: 10.1089/ars.2020.8087] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Significance: Selenoprotein P functions as a redox protein through its intrinsic thioredoxin domain and by distributing selenium to intracellular glutathione peroxidases, that is, glutathione peroxidase 1 and 4. Recent Advances: Selenoprotein P was rediscovered as a hepatokine that causes the pathology of type 2 diabetes and aging-related diseases, including exercise resistance in the skeletal muscle, insulin secretory failure in pancreatic β cells, angiogenesis resistance in vascular endothelial cells, and myocardial ischemic-reperfusion injury. It was unexpected for the antioxidant selenoprotein P to cause insulin resistance, because oxidative stress associated with obesity and fatty liver is a causal factor for hepatic insulin resistance. Critical Issues: Oxidative stress induced by the accumulation of reactive oxygen species (ROS) has a causal role in the development of insulin resistance, whereas ROS themselves function as intracellular second messengers that promote insulin signal transduction. ROS act both positively and negatively in insulin signaling depending on their concentrations. It might be possible that selenoprotein P causes "reductive stress" by eliminating a physiological ROS burst that is required for insulin signal transduction, thereby causing insulin resistance. In a large-scale intervention study, selenium supplementation that upregulates selenoprotein P was paradoxically associated with an increased risk for diabetes in humans. This review discusses the molecular mechanisms underlying the selenoprotein P-mediated resistance to angiogenesis and to exercise. Future Directions: Selenoprotein P may be the first identified intrinsic factor that induces reductive stress, causing resistance to intracellular signal transduction, which may be the therapeutic target against sedentary-lifestyle-associated diseases, such as diabetes and obesity.
Collapse
Affiliation(s)
- Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
23
|
Effects of Functionalized Fullerenes on ROS Homeostasis Determine Their Cytoprotective or Cytotoxic Properties. NANOMATERIALS 2020; 10:nano10071405. [PMID: 32707664 PMCID: PMC7407884 DOI: 10.3390/nano10071405] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/11/2020] [Accepted: 07/17/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Functionalized fullerenes (FF) can be considered regulators of intracellular reactive oxygen species (ROS) homeostasis; their direct oxidative damage-as well as regulation of oxidant enzymes and signaling pathways-should be considered. METHODS Uptake of two water-soluble functionalized C70 fullerenes with different types of aromatic addends (ethylphenylmalonate and thienylacetate) in human fetal lung fibroblasts, intracellular ROS visualization, superoxide scavenging potential, NOX4 expression, NRF2 expression, oxidative DNA damage, repair genes, cell proliferation and cell cycle were studied. RESULTS & CONCLUSION The intracellular effects of ethylphenylmalonate C70 derivative (FF1) can be explained in terms of upregulated NOX4 activity. The intracellular effects of thienylacetate C70 derivative (FF2) can be probably resulted from its superoxide scavenging potential and inhibition of lipid peroxidation. FF1 can be considered a NOX4 upregulator and potential cytotoxicant and FF2, as a superoxide scavenger and a potential cytoprotector.
Collapse
|
24
|
NADPH Oxidase 2 Mediates Myocardial Oxygen Wasting in Obesity. Antioxidants (Basel) 2020; 9:antiox9020171. [PMID: 32093119 PMCID: PMC7070669 DOI: 10.3390/antiox9020171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/03/2020] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
Obesity and diabetes are independent risk factors for cardiovascular diseases, and they are associated with the development of a specific cardiomyopathy with elevated myocardial oxygen consumption (MVO2) and impaired cardiac efficiency. Although the pathophysiology of this cardiomyopathy is multifactorial and complex, reactive oxygen species (ROS) may play an important role. One of the major ROS-generating enzymes in the cardiomyocytes is nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2), and many potential systemic activators of NOX2 are elevated in obesity and diabetes. We hypothesized that NOX2 activity would influence cardiac energetics and/or the progression of ventricular dysfunction following obesity. Myocardial ROS content and mechanoenergetics were measured in the hearts from diet-induced-obese wild type (DIOWT) and global NOK2 knock-out mice (DIOKO) and in diet-induced obese C57BL/6J mice given normal water (DIO) or water supplemented with the NOX2-inhibitor apocynin (DIOAPO). Mitochondrial function and ROS production were also assessed in DIO and DIOAPO mice. This study demonstrated that ablation and pharmacological inhibition of NOX2 both improved mechanical efficiency and reduced MVO2 for non-mechanical cardiac work. Mitochondrial ROS production was also reduced following NOX2 inhibition, while cardiac mitochondrial function was not markedly altered by apocynin-treatment. Therefore, these results indicate a link between obesity-induced myocardial oxygen wasting, NOX2 activation, and mitochondrial ROS.
Collapse
|
25
|
Wang B, Chen L, Dai L, Fang W, Wang H. Alisol B 23-Acetate Ameliorates Lipopolysaccharide-Induced Cardiac Dysfunction by Suppressing Toll-Like Receptor 4 (TLR4)/NADPH Oxidase 2 (NOX2) Signaling Pathway. Med Sci Monit 2019; 25:8472-8481. [PMID: 31707400 PMCID: PMC6863037 DOI: 10.12659/msm.918252] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Cardiac dysfunction during endotoxemia is a major cause of cardiovascular disease with high morbidity and mortality. Alisol B 23-acetate (AB23A) is a triterpenoid extracted from the Rhizoma Alismatis, a kind of traditional Chinese medicine, exhibits anti-inflammatory activity on endotoxemia. This investigation aimed to uncover the protective effects of AB23A against sepsis-induced cardiac dysfunction. Material/Methods Adult male C57BL/6 mice received lipopolysaccharide (LPS) (20 mg/kg intravenous) stimulation, with or without pre-treatment of AB23A (10 mg/kg, 20 mg/kg, or 40 mg/kg). Histopathological staining and cardiac function were performed 4 hours after LPS stimulation. Then the levels of interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α were monitored with enzyme-linked immunosorbent assay (ELISA). In addition, H9C2 cells were treated with LPS (5 μg/mL) with or without pre-treated with AB23A (0.1 μM, 1 μM, or 10 μM), and the production of reactive oxygen species (ROS) was detected by DCFH-DA combined with flow cytometry. The expression of Toll-like receptor 4 (TLR4), NADPH oxidase 2 (NOX2), NOX4, P38, p-P38, extracellular-signal-regulated kinase (ERK), and p-ERK were assessed by western blotting. Results AB23A improved the survival rate and ameliorated myocardial injury, decreased inflammatory infiltration and the level of IL-6, IL-1β, and TNF-α in the LPS-stimulated mouse model. Moreover, AB23A inhibited the ROS production in LPS-treated H9C2 cells. In addition, AB23A suppressed the levels of TLR4 and NOX2 as well as the activation levels of P38 and ERK both in vivo and in vitro. Conclusions AB23A reduced LPS-induced myocardial dysfunction by inhibiting inflammation and ROS production through the TLR4/NOX2 pathway.
Collapse
Affiliation(s)
- BinYan Wang
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Liang Chen
- Laboratory Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - LingHao Dai
- Academy of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - WenMing Fang
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| | - Hui Wang
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
26
|
Ryba DM, Warren CM, Karam CN, Davis RT, Chowdhury SAK, Alvarez MG, McCann M, Liew CW, Wieczorek DF, Varga P, Solaro RJ, Wolska BM. Sphingosine-1-Phosphate Receptor Modulator, FTY720, Improves Diastolic Dysfunction and Partially Reverses Atrial Remodeling in a Tm-E180G Mouse Model Linked to Hypertrophic Cardiomyopathy. Circ Heart Fail 2019; 12:e005835. [PMID: 31684756 DOI: 10.1161/circheartfailure.118.005835] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is a genetic cardiovascular disorder, primarily involving mutations in sarcomeric proteins. HCM patients present with hypertrophy, diastolic dysfunction, and fibrosis, but there is no specific treatment. The sphingosine-1-phosphate receptor modulator, FTY720/fingolimod, is approved for treatment of multiple sclerosis. We hypothesize that modulation of the sphingosine-1-phosphate receptor by FTY720 would be of therapeutic benefit in sarcomere-linked HCM. METHODS We treated mice with an HCM-linked mutation in tropomyosin (Tm-E180G) and nontransgenic littermates with FTY720 or vehicle for 6 weeks. Compared with vehicle-treated, FTY720-treated Tm-E180G mice had a significant reduction in left atrial size (1.99±0.19 [n=7] versus 2.70±0.44 [n=6] mm; P<0.001) and improvement in diastolic function (E/A ratio: 2.69±0.38 [n=7] versus 5.34±1.19 [n=6]; P=0.004) as assessed by echocardiography. RESULTS Pressure-volume relations revealed significant improvements in the end-diastolic pressure volume relationship, relaxation kinetics, preload recruitable stroke work, and ejection fraction. Detergent-extracted fiber bundles revealed a significant decrease in myofilament Ca2+-responsiveness (pCa50=6.15±0.11 [n=13] versus 6.24±0.06 [n=14]; P=0.041). We attributed these improvements to a downregulation of S-glutathionylation of cardiac myosin binding protein-C in FTY720-treated Tm-E180G mice and reduction in oxidative stress by downregulation of NADPH oxidases with no changes in fibrosis. CONCLUSIONS This is the first demonstration that modulation of S1PR results in decreased myofilament-Ca2+-responsiveness and improved diastolic function in HCM. We associated these changes with decreased oxidative modification of myofilament proteins via downregulation of NOX2. Our data support the hypothesis that modification of sphingolipid signaling may be a novel therapeutic approach in HCM.
Collapse
Affiliation(s)
- David M Ryba
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.)
| | - Chad M Warren
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.)
| | - Chehade N Karam
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.)
| | - Robert T Davis
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.)
| | - Shamim A K Chowdhury
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.)
| | - Manuel G Alvarez
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.)
| | - Maximilian McCann
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.)
| | - Chong Wee Liew
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.)
| | - David F Wieczorek
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, OH (D.F.W.)
| | - Peter Varga
- Department of Pediatrics, Section of Cardiology, University of Illinois at Chicago (P.V.)
| | - R John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.)
| | - Beata M Wolska
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago (D.M.R., C.M.W., C.N.K., R.T.D., S.A.K.C., M.G.A., M.M., C.W.L., R.J.S., B.M.W.).,Department of Medicine, Division of Cardiology, University of Illinois at Chicago, IL (B.M.W.)
| |
Collapse
|
27
|
Gray SP, Shah AM, Smyrnias I. NADPH oxidase 4 and its role in the cardiovascular system. ACTA ACUST UNITED AC 2019; 1:H59-H66. [PMID: 32923955 PMCID: PMC7439918 DOI: 10.1530/vb-19-0014] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 07/11/2019] [Indexed: 12/19/2022]
Abstract
The heart relies on complex mechanisms that provide adequate myocardial oxygen supply in order to maintain its contractile function. At the cellular level, oxygen undergoes one electron reduction to superoxide through the action of different types of oxidases (e.g. xanthine oxidases, uncoupled nitric oxide synthases, NADPH oxidases or NOX). Locally generated oxygen-derived reactive species (ROS) are involved in various signaling pathways including cardiac adaptation to different types of physiological and pathophysiological stresses (e.g. hypoxia or overload). The specific effects of ROS and their regulation by oxidases are dependent on the amount of ROS generated and their specific subcellular localization. The NOX family of NADPH oxidases is a main source of ROS in the heart. Seven distinct Nox isoforms (NOX1–NOX5 and DUOX1 and 2) have been identified, of which NOX1, 2, 4 and 5 have been characterized in the cardiovascular system. For the purposes of this review, we will focus on the effects of NADPH oxidase 4 (NOX4) in the heart.
Collapse
Affiliation(s)
- Stephen P Gray
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre, London, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre, London, UK
| | - Ioannis Smyrnias
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre, London, UK
| |
Collapse
|
28
|
Ronis MJ, Blackburn ML, Shankar K, Ferguson M, Cleves MA, Badger TM. Estradiol and NADPH oxidase crosstalk regulates responses to high fat feeding in female mice. Exp Biol Med (Maywood) 2019; 244:834-845. [PMID: 31161785 DOI: 10.1177/1535370219853563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We previously demonstrated protection against high fat-induced obesity in female but not male p47phox−/− mice lacking NADPH oxidase NOX1/2 activity. To test the role of estradiol (E2)-NOX crosstalk in development of this sexually dimorphic phenotype, we fed diets containing 42% fat/0.5% cholesterol to intact and ovariectomized wild type female C57BL/6 mice and female p47phox−/− mice and to ovariectomized mice where the diet was supplemented with an 1 mg/kg 17β estradiol (E2) for 12 weeks from PND28. Weight gain, gonadal fat pad weight, serum leptin and adiponectin, and adipose tissue inflammation were greater in intact wild type vs. p47 mice ( P < 0.05). Genotype effects on body weight/fat mass were abolished after ovariectomized and restored in OVX + E2 mice ( P < 0.05). The mRNA of downstream PPARγ targets CD36, lipoprotein lipase, and leptin was higher in intact wild type vs. p47phox−/− mice mice ( P < 0.05). Likewise, intact high fat-fed wild type mice had higher expression of the cytokine Mcp1; the pyroptosis marker Nirp3 and matrix remodeling and fibrosis markers Mmp2, Col1A1, and Col6a3 mRNAs ( P < 0.05). These genotype effects were reversed and restored by ovariectomized and OVX + E2, respectively ( P < 0.05). These data suggest that triglyceride accumulation in adipose tissue and development of adipose tissue injury in response to feeding diets high in fat and cholesterol is regulated by the balance between NOX-dependent reactive oxygen species signaling and E2-signaling during development. Loss of estrogens post menopause may increase the risk of obesity and metabolic syndrome as the result disinhibition of reactive oxygen species signaling. Impact statement Estrogens are known to regulate body composition. In addition, reactive oxygen species (ROS) produced by the action of NADPH oxidase (NOX) enzymes have been linked to obesity development. We examined development of obesity and adipose tissue injury in response to feeding “Western” diets high in fat and cholesterol in intact, ovariectomized (OVX), and estrogen-replaced (OVX + E2) wild type and p47phox−/− female mice where NOX2 activity is inhibited. Weight gain, gonadal fat pad weight, and adipose tissue inflammation were greater in intact WT vs. p47phox−/− mice. Genotype effects on body weight/fat mass were abolished after OVX and restored in OVX + E2 mice. These data indicate adipose tissue responses to feeding the “Western” diet is regulated by negative cross-talk between NOX-dependent ROS signaling and E2-signaling during development. Loss of estrogens post menopause may increase the risk of obesity and metabolic syndrome as the result disinhibition of ROS signaling.
Collapse
Affiliation(s)
- Martin J Ronis
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA.,3 Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Michael L Blackburn
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | - Kartik Shankar
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | - Matthew Ferguson
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | - Mario A Cleves
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | - Thomas M Badger
- 1 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,2 Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| |
Collapse
|
29
|
Li Y, Cifuentes-Pagano E, DeVallance ER, de Jesus DS, Sahoo S, Meijles DN, Koes D, Camacho CJ, Ross M, St Croix C, Pagano PJ. NADPH oxidase 2 inhibitors CPP11G and CPP11H attenuate endothelial cell inflammation & vessel dysfunction and restore mouse hind-limb flow. Redox Biol 2019; 22:101143. [PMID: 30897521 PMCID: PMC6435978 DOI: 10.1016/j.redox.2019.101143] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/05/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023] Open
Abstract
First described as essential to the phagocytic activity of leukocytes, Nox2-derived ROS have emerged as mediators of a range of cellular and tissue responses across species from salubrious to deleterious consequences. Knowledge of their role in inflammation is limited, however. We postulated that TNFα-induced endothelial reactive oxygen species (ROS) generation and pro-inflammatory signaling would be ameliorated by targeting Nox2. Herein, we in silico-modelled two first-in-class Nox2 inhibitors developed in our laboratory, explored their cellular mechanism of action and tested their efficacy in in vitro and mouse in vivo models of inflammation. Our data show that these inhibitors (CPP11G and CPP11H) disrupted canonical Nox2 organizing factor, p47phox, translocation to Nox2 in the plasma membrane; and abolished ROS production, markedly attenuated stress-responsive MAPK signaling and downstream AP-1 and NFκB nuclear translocation in human cells. Consequently, cell adhesion molecule expression and monocyte adherence were significantly inhibited by both inhibitors. In vivo, TNFα-induced ROS and inflammation were ameliorated by targeted Nox2 inhibition, which, in turn, improved hind-limb blood flow. These studies identify a proximal role for Nox2 in propagated inflammatory signaling and support therapeutic value of Nox2 inhibitors in inflammatory disease.
Collapse
Affiliation(s)
- Y Li
- Vascular Medicine Institute, USA; Department of Pharmacology & Chemical Biology, USA
| | - E Cifuentes-Pagano
- Vascular Medicine Institute, USA; Department of Pharmacology & Chemical Biology, USA
| | - E R DeVallance
- Vascular Medicine Institute, USA; Department of Pharmacology & Chemical Biology, USA
| | - D S de Jesus
- Vascular Medicine Institute, USA; Department of Pharmacology & Chemical Biology, USA
| | - S Sahoo
- Vascular Medicine Institute, USA; Department of Pharmacology & Chemical Biology, USA
| | | | - D Koes
- Computational and Systems Biology, University of Pittsburgh, USA
| | - C J Camacho
- Computational and Systems Biology, University of Pittsburgh, USA
| | - M Ross
- Center for Biologic Imaging, University of Pittsburgh, PA, 15261, USA
| | - C St Croix
- Center for Biologic Imaging, University of Pittsburgh, PA, 15261, USA
| | - P J Pagano
- Vascular Medicine Institute, USA; Department of Pharmacology & Chemical Biology, USA.
| |
Collapse
|
30
|
Diebold BA, Wilder SG, De Deken X, Meitzler JL, Doroshow JH, McCoy JW, Zhu Y, Lambeth JD. Guidelines for the Detection of NADPH Oxidases by Immunoblot and RT-qPCR. Methods Mol Biol 2019; 1982:191-229. [PMID: 31172474 DOI: 10.1007/978-1-4939-9424-3_12] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The identification of NADPH oxidase (NOX) isoforms in tissues is essential for interpreting experiments and for next step decisions regarding cell lines, animal models, and targeted drug design. Two basic methods, immunoblotting and reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR), are important to monitor NOX protein and messenger RNA (mRNA) levels, respectively, for a range of investigations from understanding cell signaling events to judging NOX inhibitor efficacies. For many other genes that are expressed in high abundance, these methods may seem rather simple. However, detecting the low expression levels of endogenous NOX/DUOX is difficult and can be frustrating, so some guidelines would be helpful to those who are facing difficulties. One reason why detection is so difficult is the limited availability of vetted NOX/DUOX antibodies. Many of the commercial antibodies do not perform well in our hands, and dependable antibodies, often generated by academic laboratories, are in limited supply. Another problem is the growing trend in the NOX literature to omit end-user validation of antibodies by not providing appropriate positive and negative controls. With regard to NOX mRNA levels, knockdown of NOX/DUOX has been reported in cell lines with very low endogenous expression (C q values ≥30) or in cell lines devoid of the targeted NOX isoform (e.g., NOX4 expression in NCI-60 cancer cell panel cell line 786-0). These publications propagate misinformation and hinder progress in understanding NOX/DUOX function. This chapter provides overdue guidelines on how to validate a NOX antibody and provides general methodologies to prepare samples for optimal detection. It also includes validated methodology to perform RT-qPCR for the measurement of NOX mRNA levels, and we suggest that RT-qPCR should be performed prior to embarking on NOX protein detection.
Collapse
Affiliation(s)
- Becky A Diebold
- Department of Pathology, Emory University, Atlanta, GA, USA.
| | | | - Xavier De Deken
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jennifer L Meitzler
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James H Doroshow
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Division of Cancer Treatment and Diagnosis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W McCoy
- Department of Pathology, Emory University, Atlanta, GA, USA
| | - Yerun Zhu
- Department of Pathology, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
31
|
Hancock M, Hafstad AD, Nabeebaccus AA, Catibog N, Logan A, Smyrnias I, Hansen SS, Lanner J, Schröder K, Murphy MP, Shah AM, Zhang M. Myocardial NADPH oxidase-4 regulates the physiological response to acute exercise. eLife 2018; 7:41044. [PMID: 30589411 PMCID: PMC6307857 DOI: 10.7554/elife.41044] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022] Open
Abstract
Regular exercise has widespread health benefits. Fundamental to these beneficial effects is the ability of the heart to intermittently and substantially increase its performance without incurring damage, but the underlying homeostatic mechanisms are unclear. We identify the ROS-generating NADPH oxidase-4 (Nox4) as an essential regulator of exercise performance in mice. Myocardial Nox4 levels increase during acute exercise and trigger activation of the transcription factor Nrf2, with the induction of multiple endogenous antioxidants. Cardiomyocyte-specific Nox4-deficient (csNox4KO) mice display a loss of exercise-induced Nrf2 activation, cardiac oxidative stress and reduced exercise performance. Cardiomyocyte-specific Nrf2-deficient (csNrf2KO) mice exhibit similar compromised exercise capacity, with mitochondrial and cardiac dysfunction. Supplementation with an Nrf2 activator or a mitochondria-targeted antioxidant effectively restores cardiac performance and exercise capacity in csNox4KO and csNrf2KO mice respectively. The Nox4/Nrf2 axis therefore drives a hormetic response that is required for optimal cardiac mitochondrial and contractile function during physiological exercise.
Collapse
Affiliation(s)
- Matthew Hancock
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Anne D Hafstad
- Cardiovascular Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Adam A Nabeebaccus
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Norman Catibog
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Angela Logan
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Ioannis Smyrnias
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Synne S Hansen
- Cardiovascular Research Group, Department of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Johanna Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologien, Goethe-Universität, Frankfurt, Germany
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Min Zhang
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| |
Collapse
|
32
|
Chandran R, Kim T, Mehta SL, Udho E, Chanana V, Cengiz P, Kim H, Kim C, Vemuganti R. A combination antioxidant therapy to inhibit NOX2 and activate Nrf2 decreases secondary brain damage and improves functional recovery after traumatic brain injury. J Cereb Blood Flow Metab 2018; 38:1818-1827. [PMID: 29083257 PMCID: PMC6168911 DOI: 10.1177/0271678x17738701] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Uncontrolled oxidative stress contributes to the secondary neuronal death that promotes long-term neurological dysfunction following traumatic brain injury (TBI). Surprisingly, both NADPH oxidase 2 (NOX2) that increases and transcription factor Nrf2 that decreases reactive oxygen species (ROS) are induced after TBI. As the post-injury functional outcome depends on the balance of these opposing molecular pathways, we evaluated the effect of TBI on the motor and cognitive deficits and cortical contusion volume in NOX2 and Nrf2 knockout mice. Genetic deletion of NOX2 improved, while Nrf2 worsened the post-TBI motor function recovery and lesion volume indicating that decreasing ROS levels might be beneficial after TBI. Treatment with either apocynin (NOX2 inhibitor) or TBHQ (Nrf2 activator) alone significantly improved the motor function after TBI, but had no effect on the lesion volume, compared to vehicle control. Whereas, the combo therapy (apocynin + TBHQ) given at either 5 min/24 h or 2 h/24 h improved motor and cognitive function and decreased cortical contusion volume compared to vehicle group. Thus, both the generation and disposal of ROS are important modulators of oxidative stress, and a combo therapy that prevents ROS formation and potentiates ROS disposal concurrently is efficacious after TBI.
Collapse
Affiliation(s)
| | - TaeHee Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Suresh L Mehta
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Eshwar Udho
- 2 Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Vishal Chanana
- 2 Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Pelin Cengiz
- 2 Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - HwuiWon Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Chanul Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,3 William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| |
Collapse
|
33
|
The role of NADPH oxidases in diabetic cardiomyopathy. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1908-1913. [DOI: 10.1016/j.bbadis.2017.07.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/19/2017] [Accepted: 07/21/2017] [Indexed: 12/14/2022]
|
34
|
Possible Modulatory Effect of Tamarind Seed Coat Extract on Fluoride-Induced Pulmonary Inflammation and Fibrosis in Rats. Inflammation 2018; 41:886-895. [DOI: 10.1007/s10753-018-0743-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Peng H, Jensen DD, Li W, Sullivan MN, Buller SA, Worker CJ, Cooper SG, Zheng S, Earley S, Sigmund CD, Feng Y. Overexpression of the neuronal human (pro)renin receptor mediates angiotensin II-independent blood pressure regulation in the central nervous system. Am J Physiol Heart Circ Physiol 2018; 314:H580-H592. [PMID: 29350998 PMCID: PMC5899258 DOI: 10.1152/ajpheart.00310.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 12/06/2017] [Accepted: 12/06/2017] [Indexed: 11/22/2022]
Abstract
Despite advances in antihypertensive therapeutics, at least 15-20% of hypertensive patients have resistant hypertension through mechanisms that remain poorly understood. In this study, we provide a new mechanism for the regulation of blood pressure (BP) in the central nervous system (CNS) by the (pro)renin receptor (PRR), a recently identified component of the renin-angiotensin system that mediates ANG II formation in the CNS. Although PRR also mediates ANG II-independent signaling, the importance of these pathways in BP regulation is unknown. Here, we developed a unique transgenic mouse model overexpressing human PRR (hPRR) specifically in neurons (Syn-hPRR). Intracerebroventricular infusion of human prorenin caused increased BP in Syn-hPRR mice. This BP response was attenuated by a NADPH oxidase (NOX) inhibitor but not by antihypertensive agents that target the renin-angiotensin system. Using a brain-targeted genetic knockdown approach, we found that NOX4 was the key isoform responsible for the prorenin-induced elevation of BP in Syn-hPRR mice. Moreover, inhibition of ERK significantly attenuated the increase in NOX activity and BP induced by human prorenin. Collectively, our findings indicate that an ANG II-independent, PRR-mediated signaling pathway regulates BP in the CNS by a PRR-ERK-NOX4 mechanism. NEW & NOTEWORTHY This study characterizes a new transgenic mouse model with overexpression of the human (pro)renin receptor in neurons and demonstrated a novel angiotensin II-independent mechanism mediated by human prorenin and the (pro)renin receptor in the central regulation of blood pressure.
Collapse
Affiliation(s)
- Hua Peng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huangzhong University of Sciences and Technology , Wuhan, Hubei , China
| | - Dane D Jensen
- Department of Physiology & Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Wencheng Li
- Department of Pathology, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Michelle N Sullivan
- Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Sophie A Buller
- Department of Physiology & Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Caleb J Worker
- Department of Physiology & Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Silvana G Cooper
- Department of Physiology & Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Shiqi Zheng
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University , Beijing , China
| | - Scott Earley
- Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Curt D Sigmund
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Yumei Feng
- Department of Physiology & Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| |
Collapse
|
36
|
Cholinergic anti-inflammatory pathway inhibits neointimal hyperplasia by suppressing inflammation and oxidative stress. Redox Biol 2017; 15:22-33. [PMID: 29197233 PMCID: PMC5723281 DOI: 10.1016/j.redox.2017.11.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/07/2017] [Accepted: 11/16/2017] [Indexed: 12/11/2022] Open
Abstract
Neointimal hyperplasia as a consequence of vascular injury is aggravated by inflammatory reaction and oxidative stress. The α7 nicotinic acetylcholine receptor (α7nAChR) is a orchestrator of cholinergic anti-inflammatory pathway (CAP), which refers to a physiological neuro-immune mechanism that restricts inflammation. Here, we investigated the potential role of CAP in neointimal hyperplasia using α7nAChR knockout (KO) mice. Male α7nAChR-KO mice and their wild-type control mice (WT) were subjected to wire injury in left common carotid artery. At 4 weeks post injury, the injured aortae were isolated for examination. The neointimal hyperplasia after wire injury was significantly aggravated in α7nAChR-KO mice compared with WT mice. The α7nAChR-KO mice had increased collagen contents and vascular smooth muscle cells (VSMCs) amount. Moreover, the inflammation was significantly enhanced in the neointima of α7nAChR-KO mice relative to WT mice, evidenced by the increased expression of tumor necrosis factor-α/interleukin-1β, and macrophage infiltration. Meanwhile, the chemokines chemokine (C-C motif) ligand 2 and chemokine (CXC motif) ligand 2 expression was also augmented in the neointima of α7nAChR-KO mice compared with WT mice. Additionally, the depletion of superoxide dismutase (SOD) and reduced glutathione (GSH), and the upregulation of 3-nitrotyrosine, malondialdehyde and myeloperoxidase were more pronounced in neointima of α7nAChR-KO mice compared with WT mice. Accordingly, the protein expression of NADPH oxidase 1 (Nox1), Nox2 and Nox4, was also higher in neointima of α7nAChR-KO mice compared with WT mice. Finally, pharmacologically activation of CAP with a selective α7nAChR agonist PNU-282987, significantly reduced neointima formation, arterial inflammation and oxidative stress after vascular injury in C57BL/6 mice. In conclusion, our results demonstrate that α7nAChR-mediated CAP is a neuro-physiological mechanism that inhibits neointima formation after vascular injury via suppressing arterial inflammation and oxidative stress. Further, these results imply that targeting α7nAChR may be a promising interventional strategy for in-stent stenosis.
Collapse
|
37
|
Mahmoud AM. Exercise Amaliorates Metabolic Disturbances and Oxidative Stress in Diabetic Cardiomyopathy: Possible Underlying Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 999:207-230. [PMID: 29022265 DOI: 10.1007/978-981-10-4307-9_12] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
38
|
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
|
39
|
Goitre L, DiStefano PV, Moglia A, Nobiletti N, Baldini E, Trabalzini L, Keubel J, Trapani E, Shuvaev VV, Muzykantov VR, Sarelius IH, Retta SF, Glading AJ. Up-regulation of NADPH oxidase-mediated redox signaling contributes to the loss of barrier function in KRIT1 deficient endothelium. Sci Rep 2017; 7:8296. [PMID: 28811547 PMCID: PMC5558000 DOI: 10.1038/s41598-017-08373-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/07/2017] [Indexed: 01/13/2023] Open
Abstract
The intracellular scaffold KRIT1/CCM1 is an established regulator of vascular barrier function. Loss of KRIT1 leads to decreased microvessel barrier function and to the development of the vascular disorder Cerebral Cavernous Malformation (CCM). However, how loss of KRIT1 causes the subsequent deficit in barrier function remains undefined. Previous studies have shown that loss of KRIT1 increases the production of reactive oxygen species (ROS) and exacerbates vascular permeability triggered by several inflammatory stimuli, but not TNF−α. We now show that endothelial ROS production directly contributes to the loss of barrier function in KRIT1 deficient animals and cells, as targeted antioxidant enzymes reversed the increase in permeability in KRIT1 heterozygous mice as shown by intravital microscopy. Rescue of the redox state restored responsiveness to TNF-α in KRIT1 deficient arterioles, but not venules. In vitro, KRIT1 depletion increased endothelial ROS production via NADPH oxidase signaling, up-regulated Nox4 expression, and promoted NF-κB dependent promoter activity. Recombinant yeast avenanthramide I, an antioxidant and inhibitor of NF-κB signaling, rescued barrier function in KRIT1 deficient cells. However, KRIT1 depletion blunted ROS production in response to TNF-α. Together, our data indicate that ROS signaling is critical for the loss of barrier function following genetic deletion of KRIT1.
Collapse
Affiliation(s)
- Luca Goitre
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Peter V DiStefano
- Department of Pharmacology and Physiology, University of Rochester, New York, USA
| | - Andrea Moglia
- Department of Agriculture, Forest and Food Sciences, Plant Genetics and Breeding, University of Torino, Torino, Italy
| | - Nicholas Nobiletti
- Department of Pharmacology and Physiology, University of Rochester, New York, USA
| | - Eva Baldini
- Department of Pharmacology and Physiology, University of Rochester, New York, USA.,Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Lorenza Trabalzini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Julie Keubel
- Department of Pharmacology and Physiology, University of Rochester, New York, USA
| | - Eliana Trapani
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Vladimir V Shuvaev
- Department of Pharmacology, University of Pennsylvania, Pennsylvania, USA
| | | | - Ingrid H Sarelius
- Department of Pharmacology and Physiology, University of Rochester, New York, USA
| | | | - Angela J Glading
- Department of Pharmacology and Physiology, University of Rochester, New York, USA.
| |
Collapse
|
40
|
Pepping JK, Vandanmagsar B, Fernandez-Kim SO, Zhang J, Mynatt RL, Bruce-Keller AJ. Myeloid-specific deletion of NOX2 prevents the metabolic and neurologic consequences of high fat diet. PLoS One 2017; 12:e0181500. [PMID: 28771483 PMCID: PMC5542654 DOI: 10.1371/journal.pone.0181500] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/03/2017] [Indexed: 12/19/2022] Open
Abstract
High fat diet-induced obesity is associated with inflammatory and oxidative signaling in macrophages that likely participates in metabolic and physiologic impairment. One key factor that could drive pathologic changes in macrophages is the pro-inflammatory, pro-oxidant enzyme NADPH oxidase. However, NADPH oxidase is a pleiotropic enzyme with both pathologic and physiologic functions, ruling out indiscriminant NADPH oxidase inhibition as a viable therapy. To determine if targeted inhibition of monocyte/macrophage NADPH oxidase could mitigate obesity pathology, we generated mice that lack the NADPH oxidase catalytic subunit NOX2 in myeloid lineage cells. C57Bl/6 control (NOX2-FL) and myeloid-deficient NOX2 (mNOX2-KO) mice were given high fat diet for 16 weeks, and subject to comprehensive metabolic, behavioral, and biochemical analyses. Data show that mNOX2-KO mice had lower body weight, delayed adiposity, attenuated visceral inflammation, and decreased macrophage infiltration and cell injury in visceral adipose relative to control NOX2-FL mice. Moreover, the effects of high fat diet on glucose regulation and circulating lipids were attenuated in mNOX2-KO mice. Finally, memory was impaired and markers of brain injury increased in NOX2-FL, but not mNOX2-KO mice. Collectively, these data indicate that NOX2 signaling in macrophages participates in the pathogenesis of obesity, and reinforce a key role for macrophage inflammation in diet-induced metabolic and neurologic decline. Development of macrophage/immune-specific NOX-based therapies could thus potentially be used to preserve metabolic and neurologic function in the context of obesity.
Collapse
Affiliation(s)
- Jennifer K. Pepping
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States of America
| | - Bolormaa Vandanmagsar
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States of America
| | - Sun-Ok Fernandez-Kim
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States of America
| | - Jingying Zhang
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States of America
| | - Randall L. Mynatt
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States of America
| | - Annadora J. Bruce-Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States of America
- * E-mail:
| |
Collapse
|
41
|
Mistry RK, Brewer AC. Redox regulation of gasotransmission in the vascular system: A focus on angiogenesis. Free Radic Biol Med 2017; 108:500-516. [PMID: 28433660 PMCID: PMC5698259 DOI: 10.1016/j.freeradbiomed.2017.04.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species have emerged as key participants in a broad range of physiological and pathophysiological processes, not least within the vascular system. Diverse cellular functions which have been attributed to some of these pro-oxidants within the vasculature include the regulation of blood pressure, neovascularisation and vascular inflammation. We here highlight the emerging roles of the enzymatically-generated reaction oxygen species, O2- and H2O2, in the regulation of the functions of the gaseous signalling molecules: nitric oxide (NO), carbon monoxide (CO), and hydrogen sulphide (H2S). These gasotransmitters are produced on demand from distinct enzymatic sources and in recent years it has become apparent that they are capable of mediating a number of homeostatic processes within the cardiovascular system including enhanced vasodilation, angiogenesis, wound healing and improved cardiac function following myocardial infarction. In common with O2- and/or H2O2 they signal by altering the functions of target proteins, either by the covalent modification of thiol groups or by direct binding to metal centres within metalloproteins, most notably haem proteins. The regulation of the enzymes which generate NO, CO and H2S have been shown to be influenced at both the transcriptional and post-translational levels by redox-dependent mechanisms, while the activity and bioavailability of the gasotransmitters themselves are also subject to oxidative modification. Within vascular cells, the family of nicotinamide adenine dinucleotide phosphate oxidases (NAPDH oxidases/Noxs) have emerged as functionally significant sources of regulated O2- and H2O2 production and accordingly, direct associations between Nox-generated oxidants and the functions of specific gasotransmitters are beginning to be identified. This review focuses on the current knowledge of the redox-dependent mechanisms which regulate the generation and activity of these gases, with particular reference to their roles in angiogenesis.
Collapse
Affiliation(s)
- Rajesh K Mistry
- Cardiovascular Division, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Alison C Brewer
- Cardiovascular Division, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK.
| |
Collapse
|
42
|
Sag CM, Schnelle M, Zhang J, Murdoch CE, Kossmann S, Protti A, Santos CXC, Sawyer G, Zhang X, Mongue-Din H, Richards DA, Brewer AC, Prysyazhna O, Maier LS, Wenzel P, Eaton PJ, Shah AM. Distinct Regulatory Effects of Myeloid Cell and Endothelial Cell NAPDH Oxidase 2 on Blood Pressure. Circulation 2017; 135:2163-2177. [PMID: 28298457 PMCID: PMC5444427 DOI: 10.1161/circulationaha.116.023877] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 03/07/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Hypertension caused by increased renin-angiotensin system activation is associated with elevated reactive oxygen species production. Previous studies implicate NADPH oxidase (Nox) proteins as important reactive oxygen species sources during renin-angiotensin system activation, with different Nox isoforms being potentially involved. Among these, Nox2 is expressed in multiple cell types, including endothelial cells, fibroblasts, immune cells, and microglia. Blood pressure (BP) is regulated at the central nervous system, renal, and vascular levels, but the cell-specific role of Nox2 in BP regulation is unknown. METHODS We generated a novel mouse model with a floxed Nox2 gene and used Tie2-Cre, LysM Cre, or Cdh5-CreERT2 driver lines to develop cell-specific models of Nox2 perturbation to investigate its role in BP regulation. RESULTS Unexpectedly, Nox2 deletion in myeloid but not endothelial cells resulted in a significant reduction in basal BP. Both Tie2-CreNox2 knockout (KO) mice (in which Nox2 was deficient in both endothelial cells and myeloid cells) and LysM CreNox2KO mice (in which Nox2 was deficient in myeloid cells) had significantly lower BP than littermate controls, whereas basal BP was unaltered in Cdh5-CreERT2 Nox2KO mice (in which Nox2 is deficient only in endothelial cells). The lower BP was attributable to an increased NO bioavailability that dynamically dilated resistance vessels in vivo under basal conditions without a change in renal function. Myeloid-specific Nox2 deletion had no effect on angiotensin II-induced hypertension, which, however, was blunted in Tie2-CreNox2KO mice, along with preservation of endothelium-dependent relaxation during angiotensin II stimulation. CONCLUSIONS We identify a hitherto unrecognized modulation of basal BP by myeloid cell Nox2, whereas endothelial cell Nox2 regulates angiotensin II-induced hypertension. These results identify distinct cell-specific roles for Nox2 in BP regulation.
Collapse
Affiliation(s)
- Can Martin Sag
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Moritz Schnelle
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Juqian Zhang
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Colin E Murdoch
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Sabine Kossmann
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Andrea Protti
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Celio X C Santos
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Greta Sawyer
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Xiaohong Zhang
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Heloise Mongue-Din
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Daniel A Richards
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Alison C Brewer
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Oleksandra Prysyazhna
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Lars S Maier
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Philip Wenzel
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Philip J Eaton
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.)
| | - Ajay M Shah
- From King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, United Kingdom (C.M.S., M.S., J.Z., C.E.M., A.P., C.X.C.S., G.S., X.Z., H.M.-D., D.A.R., A.C.B., A.P., P.J.E., A.M.S.); Klinik und Poliklinik für Innere Medizin II, Universitätsklinikum Regensburg, Regensburg, Germany (C.M.S., L.S.M.); Department of Cardiology and Pneumology, Medical Center Goettingen, Germany (M.S.); and Center for Cardiology and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Germany (S.K., P.W.).
| |
Collapse
|
43
|
Zhang X, Yang J, Yu X, Cheng S, Gan H, Xia Y. Angiotensin II-Induced Early and Late Inflammatory Responses Through NOXs and MAPK Pathways. Inflammation 2016; 40:154-165. [DOI: 10.1007/s10753-016-0464-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
44
|
Wang L, Zhao XC, Cui W, Ma YQ, Ren HL, Zhou X, Fassett J, Yang YZ, Chen Y, Xia YL, Du J, Li HH. Genetic and Pharmacologic Inhibition of the Chemokine Receptor CXCR2 Prevents Experimental Hypertension and Vascular Dysfunction. Circulation 2016; 134:1353-1368. [PMID: 27678262 PMCID: PMC5084654 DOI: 10.1161/circulationaha.115.020754] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 07/08/2016] [Indexed: 01/08/2023]
Abstract
Supplemental Digital Content is available in the text. Background: The recruitment of leukocytes to the vascular wall is a key step in hypertension development. Chemokine receptor CXCR2 mediates inflammatory cell chemotaxis in several diseases. However, the role of CXCR2 in hypertension development and the underlying mechanisms remain unknown. Methods: Angiotensin II (490 ng·kg-1·min-1) or deoxycorticosterone acetate (DOCA) salt–induced mouse hypertensive models in genetic ablation, pharmacologic inhibition of CXCR2, and adoptive bone marrow transfer mice were used to determine the role of CXCR2 in hypertension (measured by radiotelemetry and tail-cuff system), inflammation (verified by flow cytometry and quantitative real-time polymerase chain reaction [PCR] analysis), vascular remodeling (studied by haematoxylin and eosin and Masson’s trichrome staining), vascular dysfunction (assessed by aortic ring), and oxidative stress (indicated by nicotinamide adenine dinucleotide phosphate [NADPH] oxidase activity, dihydroethidium staining, and quantitative real-time PCR analysis). Moreover, the blood CXCR2+ cells in normotensive controls and hypertension patients were analyzed by flow cytometry. Results: Angiotensin II significantly upregulated the expression of CXCR2 mRNA and protein and increased the number of CD45+ CXCR2+ cells in mouse aorta (n=8 per group). Selective CXCR2 knockout (CXCR2-/-) or pharmacological inhibition of CXCR2 markedly reduced angiotensin II- or DOCA-salt-induced blood pressure elevation, aortic thickness and collagen deposition, accumulation of proinflammatory cells into the vascular wall, and expression of cytokines (n=8 per group). CXCR2 inhibition also ameliorated angiotensin II-induced vascular dysfunction and reduced vascular superoxide formation, NADPH activity, and expression of NADPH oxidase subunits (n=6 per group). Bone marrow reconstitution of wild-type mice with CXCR2-/- bone marrow cells also significantly abolished angiotensin II-induced responses (n=6 per group). It is important to note that CXCR2 blockade reversed established hypertension induced by angiotensin II or DOCA-salt challenge (n=10 per group). Furthermore, we demonstrated that CXCR2+ proinflammatory cells were higher in hypertensive patients (n=30) compared with normotensive individuals (n=20). Conclusions: Infiltration of CXCR2+ cells plays a pathogenic role in arterial hypertension and vascular dysfunction. Inhibition of CXCR2 pathway may represent a novel therapeutic approach to treat hypertension.
Collapse
Affiliation(s)
- Lei Wang
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Xue-Chen Zhao
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Wei Cui
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Yong-Qiang Ma
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Hua-Liang Ren
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Xin Zhou
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - John Fassett
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Yan-Zong Yang
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Yingjie Chen
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Yun-Long Xia
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Jie Du
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.)
| | - Hui-Hua Li
- From Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China (L.W., X.-C.Z., Y.Z.Z., Y.-L.X., H.-H.L.); Beijing Anzhen Hospital, Key Laboratory of Remodeling-Related Cardiovascular Diseases, Capital Medical University, Beijing, China (W.C., J.D.); Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China (Y.-Q.M., X.Z.); Department of Vascular Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (H.-L.R.); Department of Pharmacology and Toxicology, University of Graz, Graz, Austria (J.F.); Cardiovascular Division, University of Minnesota, Minneapolis, MN (Y.C.); and Department of Nutrition and Food Hygiene, School of Public Health, Advanced Institute of Medical Sciences, Dalian Medical University, Dalian, China (H.-H.L.).
| |
Collapse
|
45
|
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: 36] [Impact Index Per Article: 4.0] [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
|
46
|
Gharib M, Tao H, Fungwe TV, Hajri T. Cluster Differentiating 36 (CD36) Deficiency Attenuates Obesity-Associated Oxidative Stress in the Heart. PLoS One 2016; 11:e0155611. [PMID: 27195707 PMCID: PMC4873222 DOI: 10.1371/journal.pone.0155611] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/02/2016] [Indexed: 12/17/2022] Open
Abstract
RATIONALE Obesity is often associated with a state of oxidative stress and increased lipid deposition in the heart. More importantly, obesity increases lipid influx into the heart and induces excessive production of reactive oxygen species (ROS) leading to cell toxicity and metabolic dysfunction. Cluster differentiating 36 (CD36) protein is highly expressed in the heart and regulates lipid utilization but its role in obesity-associated oxidative stress is still not clear. OBJECTIVE The aim of this study was to determine the impact of CD36 deficiency on cardiac steatosis, oxidative stress and lipotoxicity associated with obesity. METHODS AND RESULTS Studies were conducted in control (Lean), obese leptin-deficient (Lepob/ob) and leptin-CD36 double null (Lepob/obCD36-/-) mice. Compared to lean mice, cardiac steatosis, and fatty acid (FA) uptake and oxidation were increased in Lepob/ob mice, while glucose uptake and oxidation was reduced. Moreover, insulin resistance, oxidative stress markers and NADPH oxidase-dependent ROS production were markedly enhanced. This was associated with the induction of NADPH oxidase expression, and increased membrane-associated p47phox, p67phox and protein kinase C. Silencing CD36 in Lepob/ob mice prevented cardiac steatosis, increased insulin sensitivity and glucose utilization, but reduced FA uptake and oxidation. Moreover, CD36 deficiency reduced NADPH oxidase activity and decreased NADPH oxidase-dependent ROS production. In isolated cardiomyocytes, CD36 deficiency reduced palmitate-induced ROS production and normalized NADPH oxidase activity. CONCLUSIONS CD36 deficiency prevented obesity-associated cardiac steatosis and insulin resistance, and reduced NADPH oxidase-dependent ROS production. The study demonstrates that CD36 regulates NADPH oxidase activity and mediates FA-induced oxidative stress.
Collapse
Affiliation(s)
- Mohamed Gharib
- Department of Surgery, Hackensack University Medical Center, New Jersey 07601, United States of America
| | - Huan Tao
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, Tennessee 37212, United States of America
| | - Thomas V. Fungwe
- Nutritional Sciences, Howard University, Washington DC 20059, United States of America
| | - Tahar Hajri
- Department of Surgery, Hackensack University Medical Center, New Jersey 07601, United States of America
- * E-mail:
| |
Collapse
|
47
|
Bórquez DA, Urrutia PJ, Wilson C, van Zundert B, Núñez MT, González-Billault C. Dissecting the role of redox signaling in neuronal development. J Neurochem 2016; 137:506-17. [DOI: 10.1111/jnc.13581] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 01/13/2016] [Accepted: 02/08/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Daniel A. Bórquez
- Facultad de Ciencias; Universidad de Chile; Santiago Chile
- Facultad de Medicina; Centro de Investigación Biomédica; Universidad Diego Portales; Santiago Chile
| | | | - Carlos Wilson
- Facultad de Ciencias; Universidad de Chile; Santiago Chile
| | | | | | - Christian González-Billault
- Facultad de Ciencias; Universidad de Chile; Santiago Chile
- Geroscience Center for Brain Health and Metabolism; Santiago Chile
| |
Collapse
|
48
|
Rezende F, Löwe O, Helfinger V, Prior KK, Walter M, Zukunft S, Fleming I, Weissmann N, Brandes RP, Schröder K. Unchanged NADPH Oxidase Activity in Nox1-Nox2-Nox4 Triple Knockout Mice: What Do NADPH-Stimulated Chemiluminescence Assays Really Detect? Antioxid Redox Signal 2016; 24:392-9. [PMID: 25906178 DOI: 10.1089/ars.2015.6314] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
NADPH oxidases of the Nox family are considered important sources of cellular reactive oxygen species (ROS) production. This conclusion is, in part, based on the ability of NADPH to elicit a chemiluminescence signal in tissue/cell homogenates or membrane preparations in the presence of enhancers such as lucigenin, luminol, or L012. However, the ability of these particular assays to specifically detect Nox activity and Nox-derived ROS has not been proven. In this study, we demonstrate that combined knockout of the three main Nox enzymes of the mouse (Nox1-Nox2-Nox4 triple knockout) had no impact on NADPH-stimulated chemiluminescence signals in the aorta, heart, and kidney homogenates. In the NADPH-stimulated membrane assays, no effect of in vivo angiotensin II pretreatment or deletion of Nox enzymes was observed. In in vitro studies in HEK293 cells, the overexpression of Nox5 or Nox4 markedly increased ROS production in intact cells, whereas overexpression of Nox5 or Nox4 had no influence on the signal in membrane assays. In contrast, overexpression of nitric oxide synthase or cytochrome P450 enzymes resulted in an increased chemiluminescence signal in isolated membranes. On the basis of these observations, we propose the hypothesis that NADPH-stimulated chemiluminescence-based membrane assays, as currently used, do not reflect Nox activity.
Collapse
Affiliation(s)
- Flávia Rezende
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany
| | - Oliver Löwe
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany
| | - Valeska Helfinger
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany
| | - Kim-Kristin Prior
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany
| | - Maria Walter
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany
| | - Sven Zukunft
- 2 Institute for Vascular Signaling, Goethe-University , Frankfurt, Germany
| | - Ingrid Fleming
- 2 Institute for Vascular Signaling, Goethe-University , Frankfurt, Germany
| | - Norbert Weissmann
- 3 Excellence Cluster Cardiopulmonary System, Justus-Liebig-University , Giessen, Germany
| | - Ralf P Brandes
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany
| | - Katrin Schröder
- 1 Institute for Cardiovascular Physiology, Goethe-University , Frankfurt, Germany
| |
Collapse
|
49
|
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
|
50
|
Santos CXC, Hafstad AD, Beretta M, Zhang M, Molenaar C, Kopec J, Fotinou D, Murray TV, Cobb AM, Martin D, Zeh Silva M, Anilkumar N, Schröder K, Shanahan CM, Brewer AC, Brandes RP, Blanc E, Parsons M, Belousov V, Cammack R, Hider RC, Steiner RA, Shah AM. Targeted redox inhibition of protein phosphatase 1 by Nox4 regulates eIF2α-mediated stress signaling. EMBO J 2016; 35:319-34. [PMID: 26742780 PMCID: PMC4741303 DOI: 10.15252/embj.201592394] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 12/02/2015] [Indexed: 01/25/2023] Open
Abstract
Phosphorylation of translation initiation factor 2α (eIF2α) attenuates global protein synthesis but enhances translation of activating transcription factor 4 (ATF4) and is a crucial evolutionarily conserved adaptive pathway during cellular stresses. The serine–threonine protein phosphatase 1 (PP1) deactivates this pathway whereas prolonging eIF2α phosphorylation enhances cell survival. Here, we show that the reactive oxygen species‐generating NADPH oxidase‐4 (Nox4) is induced downstream of ATF4, binds to a PP1‐targeting subunit GADD34 at the endoplasmic reticulum, and inhibits PP1 activity to increase eIF2α phosphorylation and ATF4 levels. Other PP1 targets distant from the endoplasmic reticulum are unaffected, indicating a spatially confined inhibition of the phosphatase. PP1 inhibition involves metal center oxidation rather than the thiol oxidation that underlies redox inhibition of protein tyrosine phosphatases. We show that this Nox4‐regulated pathway robustly enhances cell survival and has a physiologic role in heart ischemia–reperfusion and acute kidney injury. This work uncovers a novel redox signaling pathway, involving Nox4–GADD34 interaction and a targeted oxidative inactivation of the PP1 metal center, that sustains eIF2α phosphorylation to protect tissues under stress.
Collapse
Affiliation(s)
- Celio X C Santos
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Anne D Hafstad
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK Cardiovascular Research Group, Department of Medical Biology, The Arctic University of Norway, Tromsø, Norway
| | - Matteo Beretta
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Min Zhang
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Chris Molenaar
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Jola Kopec
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK Randall Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Dina Fotinou
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK Randall Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Thomas V Murray
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Andrew M Cobb
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Daniel Martin
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Maira Zeh Silva
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK Randall Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Narayana Anilkumar
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany
| | - Catherine M Shanahan
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Alison C Brewer
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany
| | - Eric Blanc
- MRC Centre for Developmental Neurobiology, King's College London, London, UK
| | - Maddy Parsons
- Randall Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Vsevelod Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Richard Cammack
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Robert C Hider
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Roberto A Steiner
- Randall Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ajay M Shah
- Cardiovascular Division, King's College London British Heart Foundation Centre of Excellence, London, UK
| |
Collapse
|