1
|
Zare F, Ghafouri-Fard S, Shamosi A, Pahlavan S, Mahboudi H, Tavasoli A, Eslami S. Oleoylethanolamide protects mesenchymal stem/stromal cells (MSCs) from oxidative stress and reduces adipogenic related genes expression in adipose-derived MSCs undergoing adipocyte differentiation. Mol Biol Rep 2023; 51:33. [PMID: 38155334 DOI: 10.1007/s11033-023-08929-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/24/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Human mesenchymal stem/stromal cells (hMSCs) are known for their pronounced therapeutic potential; however, they are still applied in limited clinical cases for several reasons. ROS-mediated oxidative stress is among the chief causes of post-transplantation apoptosis and death of hMSCs. It has been reported that a strategy to protect hMSCs against ROS is to pretreat them with antioxidants. Oleoylethanolamide (OEA) is a monounsaturated fatty acid derived from oleic acid and it has many protective properties, including anti-obesity, anti-inflammatory, and antioxidant effects. OEA is also used as a weight loss supplement; due to its high affinity for the PPAR-α receptor, OEA increases the fat metabolism rate. METHODS AND RESULTS This study hence assessed the effects of OEA pretreatment on the in vitro survival rate and resistance of hMSCs under oxidative stress as well as the cellular and molecular events in the biology of stem/stromal cells affected by oxidative stress and free radicals. Considering the role of MSCs in adipogenesis and obesity, the expression of the main genes involved in adipogenesis was also addressed in this study. Results revealed that OEA increases the in vitro proliferation of MSCs and inhibits cell apoptosis by reducing the induction of oxidative stress. The results also indicated that OEA exerts its antioxidant properties by both activating the Nrf2/NQO-1/HO-1 signaling pathway and directly combating free radicals. Moreover, OEA can reduce adipogenesis through reducing the expression of PPARγ, leptin and CEBPA genes in hMSCs undergoing adipocyte differentiation. CONCLUSIONS Thus, OEA protects hMSCs from oxidative stress and reduces adipogenic related genes expression and can be regarded as a therapeutic agent for this purpose.
Collapse
Affiliation(s)
- Fereshteh Zare
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Soudeh Ghafouri-Fard
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefeh Shamosi
- Department of Anatomy, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Shahrzad Pahlavan
- Department of Anatomy, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Hossein Mahboudi
- Department of Biotechnology, Faculty of Pharmacy, Alborz University of Medical Sciences, Karaj, Iran
| | - Afsaneh Tavasoli
- Department of Biotechnology, Faculty of Pharmacy, Alborz University of Medical Sciences, Karaj, Iran
| | - Solat Eslami
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran.
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
2
|
Han X, Wang H, Du F, Zeng X, Guo C. Nrf2 for a key member of redox regulation: A novel insight against myocardial ischemia and reperfusion injuries. Biomed Pharmacother 2023; 168:115855. [PMID: 37939614 DOI: 10.1016/j.biopha.2023.115855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023] Open
Abstract
Nuclear factor erythroid-2 related factor 2 (Nrf2), a nuclear transcription factor, modulates genes responsible for antioxidant responses against toxic and oxidative stress to maintain redox homeostasis and participates in varieties of cellular processes such as metabolism and inflammation during myocardial ischemia and reperfusion injuries (MIRI). The accumulation of reactive oxygen species (ROS) from damaged mitochondria, xanthine oxidase, NADPH oxidases, and inflammation contributes to depraved myocardial ischemia and reperfusion injuries. Considering that Nrf2 played crucial roles in antagonizing oxidative stress, it is reasonable to delve into the up or down-regulated molecular mechanisms of Nrf2 in the progression of MIRI to provide the possibility of new therapeutic medicine targeting Nrf2 in cardiovascular diseases. This review systematically describes the generation of ROS, the regulatory metabolisms of Nrf2 as well as several natural or synthetic compounds activating Nrf2 during MIRI, which might provide novel insights for the anti-oxidative stress and original ideas targeting Nrf2 for the prevention and treatment in cardiovascular diseases.
Collapse
Affiliation(s)
- Xuejie Han
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China
| | - Hongxia Wang
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China
| | - Fenghe Du
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing 100070, PR China
| | - Xiangjun Zeng
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China.
| | - Caixia Guo
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China.
| |
Collapse
|
3
|
Mariappan V, Adikari S, Shanmugam L, Easow JM, Balakrishna Pillai A. Differential expression of interferon inducible protein: Guanylate binding protein (GBP1 & GBP2) in severe dengue. Free Radic Biol Med 2023; 194:131-146. [PMID: 36460216 DOI: 10.1016/j.freeradbiomed.2022.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/17/2022] [Accepted: 11/27/2022] [Indexed: 12/02/2022]
Abstract
Dengue virus is reported to activate endothelial cells (EC), but the precise cause for severe dengue (SD) is not known. Guanylate binding proteins (GBPs) are IFN-inducible proteins secreted by ECs and are involved in the anti-oxidant and anti-viral response. The involvement of GBPs in the pathogenesis of dengue remains under explored. In the present study, we quantified the mRNA and protein levels of GBP1 and 2 during acute, defervescence and convalescent phase in SD-10, dengue without warning sign-15 and dengue with warning sign-25 compared to other febrile illnesses-10 and healthy controls-8 using RT-PCR and ELISA respectively. Lipid peroxidation in plasma samples were measured using the Kei Satoh method. Protein and DNA oxidation were determined by ELISA. The efficacy of the proteins in predicting disease severity was done by Support Vector Machine (SVM) model. A significant (P ≤ 0.01) decrease in the levels of mRNA and protein of both GBP1 and GBP2 was observed during defervescence in both SD and DWW cases. The levels were significantly (P ≤ 0.05) tapered off in SD cases from acute till critical phases compared to other study groups. DNA, protein and lipid oxidation markers showed an increasing trend in SD (P ≤ 0.01). Both GBP1 & 2 were found to be negatively associated plasma leakage and oxidative stress markers. EC's activated with SD serum showed a reduced expression of GBP 1 and 2. Nevertheless, the SVM model revealed that plasma levels of proteins along with clinical symptoms could predict the disease outcomes with higher precision. This is the first study reporting a downregulated expression of GBP1 & 2 and their association with oxidative stress and plasma leakage in dengue cases. This suggests the importance of GBPs in regulating disease manifestation. However, further investigations are required to ascertain its role as a biomarker or therapeutic target in dengue infection.
Collapse
Affiliation(s)
- Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI) (Formerly Central Inter-Disciplinary Research Facility-CIDRF), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India
| | - Shalinda Adikari
- Department of Information System and Analytics, National University of Singapore (NUS), Singapore, 117 417, Republic of Singapore
| | - Lokesh Shanmugam
- ICMR-National Institute of Epidemiology (ICMR-NIE), Ayapakkam, Chennai, 600 070, India; Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India
| | - Joshy M Easow
- Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India
| | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI) (Formerly Central Inter-Disciplinary Research Facility-CIDRF), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607 402, India.
| |
Collapse
|
4
|
Ishii T, Warabi E, Mann GE. Mechanisms underlying Nrf2 nuclear translocation by non-lethal levels of hydrogen peroxide: p38 MAPK-dependent neutral sphingomyelinase2 membrane trafficking and ceramide/PKCζ/CK2 signaling. Free Radic Biol Med 2022; 191:191-202. [PMID: 36064071 DOI: 10.1016/j.freeradbiomed.2022.08.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 12/14/2022]
Abstract
Hydrogen peroxide is an aerobic metabolite playing a central role in redox signaling and oxidative stress. H2O2 could activate redox sensitive transcription factors, such as Nrf2, AP-1 and NF-κB by different manners. In some cells, treatment with non-lethal levels of H2O2 induces rapid activation of Nrf2, which upregulates expression of a set of genes involved in glutathione (GSH) synthesis and defenses against oxidative damage. It depends on two steps, the rapid translational activation of Nrf2 and facilitation of Nrf2 nuclear translocation. We review the molecular mechanisms by which H2O2 induces nuclear translocation of Nrf2 in cultured cells by highlighting the role of neutral sphingomyelinase 2 (nSMase2), a GSH sensor. H2O2 enters cells through aquaporin channels in the plasma membrane and is rapidly reduced to H2O by GSH peroxidases to consume cellular GSH, resulting in nSMase2 activation to generate ceramide. H2O2 also activates p38 MAP kinase, which enhances transfer of nSMase2 from perinuclear regions to plasma membrane lipid rafts to accelerate ceramide generation. Low levels of ceramide activate PKCζ, which then activates casein kinase 2 (CK2). These protein kinases are able to phosphorylate Nrf2 to stabilize and activate it. Notably, Nrf2 also binds to caveolin-1 (Cav1), which protects Nrf2 from Keap1-mediated degradation and limits Nrf2 nuclear translocation. We propose that Cav1serves as a signaling hub for the control of H2O2-mediated phosphorylation of Nrf2 by kinases, which results in release of Nrf2 from Cav1 to facilitate nuclear translocation. In summary, H2O2 induces GSH depletion which is recovered by Nrf2 activation dependent on p38/nSMase2/ceramide signaling.
Collapse
Affiliation(s)
- Tetsuro Ishii
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Eiji Warabi
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
5
|
Akki R, Siracusa R, Cordaro M, Remigante A, Morabito R, Errami M, Marino A. Adaptation to oxidative stress at cellular and tissue level. Arch Physiol Biochem 2022; 128:521-531. [PMID: 31835914 DOI: 10.1080/13813455.2019.1702059] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Several in vitro and in vivo investigations have already proved that cells and tissues, when pre-exposed to low oxidative stress by different stimuli such as chemical, physical agents and environmental factors, display more resistance against subsequent stronger ischaemic injuries, resulting in an adaptive response known as ischaemic preconditioning (IPC). The aim of this review is to report the most recent knowledge about the complex adaptive mechanisms, including signalling transduction pathways, antioxidant systems, apoptotic and inflammation pathways, underlying cell protection against oxidative damage. In addition, an update about in vivo adaptation strategies in response to ischaemic/reperfusion episodes and brain trauma is also given.
Collapse
Affiliation(s)
- Rachid Akki
- Department of Biology, Faculty of Science, University of Abdelmalek Essaadi, Tetouan, Morocco
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Remigante
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Rossana Morabito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Mohammed Errami
- Department of Biology, Faculty of Science, University of Abdelmalek Essaadi, Tetouan, Morocco
| | - Angela Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
6
|
Modification of Ischemia/Reperfusion-Induced Alterations in Subcellular Organelles by Ischemic Preconditioning. Int J Mol Sci 2022; 23:ijms23073425. [PMID: 35408783 PMCID: PMC8998910 DOI: 10.3390/ijms23073425] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/07/2023] Open
Abstract
It is now well established that ischemia/reperfusion (I/R) injury is associated with the compromised recovery of cardiac contractile function. Such an adverse effect of I/R injury in the heart is attributed to the development of oxidative stress and intracellular Ca2+-overload, which are known to induce remodeling of subcellular organelles such as sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils. However, repeated episodes of brief periods of ischemia followed by reperfusion or ischemic preconditioning (IP) have been shown to improve cardiac function and exert cardioprotective actions against the adverse effects of prolonged I/R injury. This protective action of IP in attenuating myocardial damage and subcellular remodeling is likely to be due to marked reductions in the occurrence of oxidative stress and intracellular Ca2+-overload in cardiomyocytes. In addition, the beneficial actions of IP have been attributed to the depression of proteolytic activities and inflammatory levels of cytokines as well as the activation of the nuclear factor erythroid factor 2-mediated signal transduction pathway. Accordingly, this review is intended to describe some of the changes in subcellular organelles, which are induced in cardiomyocytes by I/R for the occurrence of oxidative stress and intracellular Ca2+-overload and highlight some of the mechanisms for explaining the cardioprotective effects of IP.
Collapse
|
7
|
Zhu C, Gu H, Jin Y, Wurm D, Freidhof B, Lu Y, Chen QM. Metabolomics of oxidative stress: Nrf2 independent depletion of NAD or increases of sugar alcohols. Toxicol Appl Pharmacol 2022; 442:115949. [PMID: 35227738 DOI: 10.1016/j.taap.2022.115949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 01/07/2023]
Abstract
Nrf2 encodes a transcription factor best known for regulating the expression of antioxidant and detoxification genes. Recent evidence suggested that Nrf2 mediates metabolic reprogramming in cancer cells. However, the role of Nrf2 in the biochemical metabolism of cardiac cells has not been studied. Using LC-MS/MS-based metabolomics, we addressed whether knocking out the Nrf2 gene in AC16 human cardiomyocytes affects metabolic reprogramming by oxidative stress. Profiling the basal level metabolites showed an elevated pentose phosphate pathway and increased levels of sugar alcohols, sorbitol, L-arabitol, xylitol and xylonic acid, in Nrf2 KO cells. With sublethal levels of oxidative stress, depletion of NAD, an increase of GDP and elevation of sugar alcohols, sorbitol and dulcitol, were detected in parent wild type (WT) cells. Knocking out Nrf2 did not affect these changes. Biochemical assays confirmed depletion of NAD in WT and Nrf2 KO cells due to H2O2 treatment. These data support that although Nrf2 deficiency caused baseline activation of the pentose phosphate pathway and sugar alcohol synthesis, a brief exposure to none-lethal doses of H2O2 caused NAD depletion in an Nrf2 independent manner. Loss of NAD may contribute to oxidative stress associated cell degeneration as observed with aging, diabetes and heart failure.
Collapse
|
8
|
Chen QM. Nrf2 for cardiac protection: pharmacological options against oxidative stress. Trends Pharmacol Sci 2021; 42:729-744. [PMID: 34332753 DOI: 10.1016/j.tips.2021.06.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 01/07/2023]
Abstract
Myocardial ischemia or reperfusion increases the generation of reactive oxygen species (ROS) from damaged mitochondria, NADPH oxidases, xanthine oxidase, and inflammation. ROS can be removed by eight endogenous antioxidant and redox systems, many components of which are expressed under the influence of the activated Nrf2 transcription factor. Transcriptomic profiling, sequencing of Nrf2-bound DNA, and Nrf2 gene knockout studies have revealed the power of Nrf2 beyond the antioxidant and detoxification response, from tissue recovery, repair, and remodeling, mitochondrial turnover, and metabolic reprogramming to the suppression of proinflammatory cytokines. Multifaceted regulatory mechanisms for Nrf2 protein levels or activity have been mapped to its functional domains, Nrf2-ECH homology (Neh)1-7. Oxidative stress activates Nrf2 via nuclear translocation, de novo protein translation, and increased protein stability due to removal of the Kelch-like ECH-associated protein 1 (Keap1) checkpoint, or the inactivation of β-transducin repeat-containing protein (β-TrCP), or Hmg-CoA reductase degradation protein 1 (Hrd1). The promise of small-molecule Nrf2 inducers from natural products or derivatives is discussed here. Experimental evidence is presented to support Nrf2 as a lead target for drug development to further improve the treatment outcome for myocardial infarction (MI).
Collapse
Affiliation(s)
- Qin M Chen
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
9
|
Wang J, Zhai Y, Ou M, Bian Y, Tang C, Zhang W, Cheng Y, Li G. Protective Effect of Lemon Peel Extract on Oxidative Stress in H9c2 Rat Heart Cell Injury. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2047-2058. [PMID: 34017169 PMCID: PMC8131012 DOI: 10.2147/dddt.s304624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/21/2021] [Indexed: 01/27/2023]
Abstract
Aim Lemon peel, a traditional Chinese medicine, was tested in this study for its novel application in inhibiting cellular oxidative stress, and the effect of lemon peel extract (LPE) on protecting H9c2 rat heart cells from oxidative stress was investigated. Methods The scavenging effects of LPE on 1,1-diphenyl-2-picryhydrazyl (DPPH) and 2,2’-azino-bis (3-ethylbenzthiazoline-6-sulfonic acid) (ABTS) free radicals were measured in extracellular experiments. The 3-(4,5-dimethyl-2-thiazolinyl)-2,5-diphenyl-2-h-tetrazolylammonium bromide (MTT) assay was used to detect the cell survival rate. The cell supernatant and intracellular oxidation-related indicators were detected by a kit, and the mRNA expression in H9c2 cells was detected by quantitative polymerase chain reaction (qPCR). The chemical substances of LPE were analyzed by high-performance liquid chromatography (HPLC). Results The results showed that LPE exhibited better DPPH and ABTS free radical scavenging abilities than vitamin C. Compared with the cells in the normal state (control group), the cell survival rate in the model group decreased, and the level of lactate dehydrogenase (LDH) increased, the levels of superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) decreased, and the content of malondialdehyde (MDA) increased. Compared with the control group, the expression of Bcl-2-related X protein (Bax), caspase-3, nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) in the model group was increased, and the expression of B-cell lymphoma-2 (Bcl-2) was reduced. Compared with the model group, LPE treatment improved the cell survival rate, reduced the levels of LDH and MDA, increased the levels of SOD, CAT, and GSH, downregulated the expression of Bax, caspase-3, Nrf2 and HO-1, and upregulated the expression of Bcl-2. The composition analysis showed that LPE contained catechin, rutin, naringin, quercetin, and hesperidin. Conclusion The results indicated that LPE could protect H9c2 cells from oxidative stress through five active components. LPE has the potential to be developed into natural medicine or health food for the inhibition of cell oxidative damage.
Collapse
Affiliation(s)
- Jun Wang
- Citrus Research Institute, Southwest University, Chongqing, People's Republic of China.,National Citrus Engineering Research Center, Chongqing, People's Republic of China
| | - Yulin Zhai
- Citrus Research Institute, Southwest University, Chongqing, People's Republic of China
| | - Mingguang Ou
- Guang'an Nongfeng Agricultural Development Co., Ltd, Sichuan, People's Republic of China
| | - Yunfeng Bian
- Guang'an Zheng Wang Agriculture Co., Ltd, Sichuan, People's Republic of China
| | - Chenglong Tang
- Yuanyang Hongtu Grapefruit Agricultural Technology Development Co., Ltd, Yunnan, People's Republic of China
| | - Wanchao Zhang
- Chongqing Institute of Medicinal Plant Cultivation, Chongqing, People's Republic of China.,National Patent Navigation Project (Chongqing) Research and Promotion Center, Chongqing, People's Republic of China.,Bio-Resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chongqing, People's Republic of China
| | - Yujiao Cheng
- Citrus Research Institute, Southwest University, Chongqing, People's Republic of China.,National Citrus Engineering Research Center, Chongqing, People's Republic of China
| | - Guijie Li
- Citrus Research Institute, Southwest University, Chongqing, People's Republic of China.,National Citrus Engineering Research Center, Chongqing, People's Republic of China
| |
Collapse
|
10
|
Harnessing the cardiovascular benefits of exercise: are Nrf2 activators useful? SPORTS MEDICINE AND HEALTH SCIENCE 2021; 3:70-79. [PMID: 35782161 PMCID: PMC9219337 DOI: 10.1016/j.smhs.2021.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 01/07/2023] Open
Abstract
The ability of physical activity to ameliorate cardiovascular disease and improve cardiovascular health is well accepted, but many aspects of the molecular mechanisms underlying these benefits are incompletely understood. Exercise increases the levels of reactive oxygen species (ROS) through various mechanisms. This triggers the activation of Nrf2, a redox-sensitive transcription factor activated by increases in oxidative stress. Activation of Nrf2 mitigates oxidative stress by increasing the nuclear transcription of many antioxidant genes while also mediating additional beneficial effects through the cytoprotective nature of Nrf2 signaling. Understanding the transcriptional patterns of Nrf2 caused by exercise can help in the design of pharmacological mimicry of the process in patients who are unable to exercise for various reasons.
Collapse
|
11
|
Heat Shock Proteins in Oxidative Stress and Ischemia/Reperfusion Injury and Benefits from Physical Exercises: A Review to the Current Knowledge. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6678457. [PMID: 33603951 PMCID: PMC7868165 DOI: 10.1155/2021/6678457] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
Heat shock proteins (HSPs) are molecular chaperones produced in response to oxidative stress (OS). These proteins are involved in the folding of newly synthesized proteins and refolding of damaged or misfolded proteins. Recent studies have been focused on the regulatory role of HSPs in OS and ischemia/reperfusion injury (I/R) where reactive oxygen species (ROS) play a major role. ROS perform many functions, including cell signaling. Unfortunately, they are also the cause of pathological processes leading to various diseases. Biological pathways such as p38 MAPK, HSP70 and Akt/GSK-3β/eNOS, HSP70, JAK2/STAT3 or PI3K/Akt/HSP70, and HSF1/Nrf2-Keap1 are considered in the relationship between HSP and OS. New pathophysiological mechanisms involving ROS are being discovered and described the protein network of HSP interactions. Understanding of the mechanisms involved, e.g., in I/R, is important to the development of treatment methods. HSPs are multifunctional proteins because they closely interact with the antioxidant and the nitric oxide generation systems, such as HSP70/HSP90/NOS. A deficiency or excess of antioxidants modulates the activation of HSF and subsequent HSP biosynthesis. It is well known that HSPs are involved in the regulation of several redox processes and play an important role in protein-protein interactions. The latest research focuses on determining the role of HSPs in OS, their antioxidant activity, and the possibility of using HSPs in the treatment of I/R consequences. Physical exercises are important in patients with cardiovascular diseases, as they affect the expression of HSPs and the development of OS.
Collapse
|
12
|
Beneficial effects of Diplectria barbata (Wall. Ex C. B. Clarke) Franken et Roos extract on aging and antioxidants in vitro and in vivo. Toxicol Res 2020; 37:71-83. [PMID: 33489859 DOI: 10.1007/s43188-020-00064-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 01/02/2023] Open
Abstract
The purpose of this study is to explore the effects of Diplectria barbata (Wall. Ex C.B. Clarke) Franken & Roons (DFR) on wound healing, antioxidant and aging in Normal Human Dermal Fibroblast cell (NHDF) cells and mouse skin models. We investigated the effects of the aging process in vitro and in vivo. DFRtreated NHDF cells showed a concentration-dependent increase in the expression of extracellular matrix (ECM) proteins (Collagen-2.5-fold increase at 50 μg/ml, Elastin-1.5-fold increase at 1μg/ml) as well as an increase in proteins related to cell survival, differentiation, and development, while expression of aging proteins such as matrix metalloproteinase 3 (MMP-3) was decreased (5-fold decrease at 50 μg/ml). DFR treatment also led to enhanced expression of antioxidant proteins such as nuclear factor erythroid 2-related factor 2 (10-fold increase at 50 μg/ml) and heme oxygenase 1 (1.5-fold increase at 25 μg/ml). To further investigate the antioxidative effects of DFR extracts, the 2,2-diphenyl-1-picrylhydrazyl radical scavenging activities were also evaluated. DFR extracts improved wound healing and resulted in increased expression of ECM proteins, while enzymes involved in collagen degradation, including MMP-3, were decreased in NHDF cells as well as in a mouse model. This study demonstrates the anti-aging, antioxidant, and wound healing properties of DFR extracts. Therefore, DFR extracts present may facilitate skin protection and care.
Collapse
|
13
|
Shemarova IV, Korotkov SM, Nesterov VP. Ca2+-Dependent
Mitochondrial Mechanisms of Cardioprotection. J EVOL BIOCHEM PHYS+ 2020. [DOI: 10.1134/s002209302004002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
14
|
Loxham M, Woo J, Singhania A, Smithers NP, Yeomans A, Packham G, Crainic AM, Cook RB, Cassee FR, Woelk CH, Davies DE. Upregulation of epithelial metallothioneins by metal-rich ultrafine particulate matter from an underground railway. Metallomics 2020; 12:1070-1082. [PMID: 32297622 DOI: 10.1039/d0mt00014k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Airborne particulate matter (PM) is a leading cause of mortality and morbidity. However, understanding of the range and mechanisms of effects of PM components is poor. PM generated in underground railways is rich in metals, especially iron. In the ultrafine (UFPM; <0.1 μm diameter) fraction, the combination of small size and metal enrichment poses an unknown health risk. This study aimed to analyse transcriptomic responses to underground UFPM in primary bronchial epithelial cells (PBECs), a key site of PM deposition. The oxidation state of iron in UFPM from an underground station was determined by X-ray absorption near edge structure (XANES) spectroscopy. Antioxidant response was assayed using a reporter cell line transfected with an antioxidant response element (ARE)-luciferase construct. Differentiated PBECs were exposed to UFPM for 6 h or 24 h for RNA-Seq and RT-qPCR analysis. XANES showed predominance of redox-active Fe3O4, with ROS generation confirmed by induction of ARE-luciferase expression. 6 h exposure of PBECs to UFPM identified 52 differentially expressed genes (DEGs), especially associated with epithelial maintenance, whereas 24 h exposure yielded 23 DEGs, particularly involved with redox homeostasis and metal binding. At both timepoints, there was upregulation of members of the metallothionein family, low molecular weight proteins with antioxidant activity whose main function is binding and homeostasis of zinc and copper ions, but not iron ions. This upregulation was partially inhibited by metal chelation or ROS scavenging. These data suggest differential regulation of responses to metal-rich UFPM depending on exposure period, and highlight novel pathways and markers of PM exposure, with the role of metallothioneins warranting further investigation.
Collapse
Affiliation(s)
- Matthew Loxham
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Tremona Road, Southampton, UKSO16 6YD. and NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Tremona Road, Southampton, UKSO16 6YD and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton, UKSO17 1BJ and Southampton Marine and Maritime Institute, University of Southampton, Boldrewood Innovation Campus, Southampton, UKSO16 7QF
| | - Jeongmin Woo
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Tremona Road, Southampton, UKSO16 6YD.
| | - Akul Singhania
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Tremona Road, Southampton, UKSO16 6YD.
| | - Natalie P Smithers
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Tremona Road, Southampton, UKSO16 6YD.
| | - Alison Yeomans
- Cancer Research UK Centre, Cancer Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UKSO16 6YD
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Southampton, UKSO16 6YD
| | - Alina M Crainic
- National Centre for Advanced Tribology (nCATS), Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UKSO17 1BJ
| | - Richard B Cook
- National Centre for Advanced Tribology (nCATS), Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UKSO17 1BJ
| | - Flemming R Cassee
- Centre for Sustainability, Environment, and Health, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands and Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, The Netherlands
| | - Christopher H Woelk
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Tremona Road, Southampton, UKSO16 6YD.
| | - Donna E Davies
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, University Hospital Southampton, Tremona Road, Southampton, UKSO16 6YD. and NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Tremona Road, Southampton, UKSO16 6YD and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton, UKSO17 1BJ
| |
Collapse
|
15
|
Bauman BM, Jeong C, Savage M, Briker AL, Janigian NG, Nguyen LL, Kemmerer ZA, Eggler AL. Dr. Jekyll and Mr. Hyde: Oxidizable phenol-generated reactive oxygen species enhance sulforaphane's antioxidant response element activation, even as they suppress Nrf2 protein accumulation. Free Radic Biol Med 2018; 124:532-540. [PMID: 29969714 DOI: 10.1016/j.freeradbiomed.2018.06.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/17/2022]
Abstract
The transcription factor Nrf2 is a master regulator of antioxidant and cytoprotective genes, binding to antioxidant response elements (AREs) in their promoter regions. Due to the therapeutic role of the Nrf2/ARE system in oxidative homeostasis, its activation has been investigated in many pre-clinical and clinical trials for common chronic diseases. One of the most promising Nrf2 activators is sulforaphane, the subject of over 50 clinical trials. In this work, we examine the effect of reactive oxygen species (ROS) on sulforaphane's Nrf2/ARE activation in the non-tumorigenic keratinocyte cell line HaCaT, with the non-arylating oxidizable phenol, 2,5-di-tert-butylhydroquinone (dtBHQ), as the source of ROS. We find that, in combination with 2.5 µM sulforaphane, dtBHQ markedly enhances ARE-regulated gene expression, including expression of the cytoprotective proteins aldo-keto reductase family 1 member C1 (AKR1C1) and heme oxygenase-1 (HO-1). Additionally, sulforaphane's therapeutic window is widened by 12.5 µM dtBHQ. Our data suggest that H2O2 generated by dtBHQ oxidation is responsible for these effects, as shown by inclusion of catalase and by co-treatment with sulforaphane and H2O2. While sulforaphane treatment causes Nrf2 protein to accumulate as expected, interestingly, dtBHQ and H2O2 appear to act on targets downstream of Nrf2 protein accumulation to enhance sulforaphane's ARE-regulated gene expression. Inclusion of dtBHQ or H2O2 with sulforaphane does not increase Nrf2 protein levels, and catalase has little effect on Nrf2 protein levels in the presence of sulforaphane and dtBHQ. Surprisingly, dtBHQ suppresses Nrf2 protein synthesis. Inclusion of a superoxide dismutase mimetic with sulforaphane and dtBHQ partly rescues Nrf2 suppression and significantly further increases sulforaphane's efficacy for ARE-reporter expression. Thus, there is a "Dr. Jekyll and Mr. Hyde" effect of ROS: ROS enhance sulforaphane's ARE-regulated gene expression even as they also inhibit Nrf2 protein synthesis. This unexpected finding reveals the degree to which targets in the ARE pathway downstream of Nrf2 protein accumulation contribute to gene expression. The results presented here provide a model system for significant enhancement of sulforaphane's potency with small molecule co-treatment.
Collapse
Affiliation(s)
- Bradly M Bauman
- Department of Chemistry, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085, United States
| | - Chang Jeong
- Department of Chemistry, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085, United States
| | - Matthew Savage
- Department of Chemistry, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085, United States
| | - Anna L Briker
- Department of Chemistry, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085, United States
| | - Nicholas G Janigian
- Department of Chemistry, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085, United States
| | - Linda L Nguyen
- Department of Chemistry, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085, United States
| | - Zachary A Kemmerer
- Department of Chemistry, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085, United States
| | - Aimee L Eggler
- Department of Chemistry, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085, United States.
| |
Collapse
|
16
|
Lee H, Hong Y, Tran Q, Cho H, Kim M, Kim C, Kwon SH, Park S, Park J, Park J. A new role for the ginsenoside RG3 in antiaging via mitochondria function in ultraviolet-irradiated human dermal fibroblasts. J Ginseng Res 2018; 43:431-441. [PMID: 31308815 PMCID: PMC6606973 DOI: 10.1016/j.jgr.2018.07.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/09/2018] [Indexed: 11/18/2022] Open
Abstract
Background The efficacy of ginseng, the representative product of Korea, and its chemical effects have been well investigated. The ginsenoside RG3 has been reported to exhibit apoptotic, anticancer, and antidepressant-like effects. Methods In this report, the putative effect of RG3 on several cellular function including cell survival, differentiation, development and aging process were evaluated by monitoring each specific marker. Also, mitochondrial morphology and function were investigated in ultraviolet (UV)-irradiated normal human dermal fibroblast cells. Results RG3 treatment increased the expression of extracellular matrix proteins, growth-associated immediate-early genes, and cell proliferation genes in UV-irradiated normal human dermal fibroblast cells. And, RG3 also resulted in enhanced expression of antioxidant proteins such as nuclear factor erythroid 2–related factor-2 and heme oxygenase-1. In addition, RG3 affects the morphology of UV-induced mitochondria and plays a role in protecting mitochondrial dysfunction. Conclusioin RG3 restores mitochondrial adenosine triphosphate (ATP) and membrane potential via its antioxidant effects in skin cells damaged by UV irradiation, leading to an increase in proteins linked with the extracellular matrix, cell proliferation, and antioxidant activity.
Collapse
Affiliation(s)
- Hyunji Lee
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Youngeun Hong
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Quangdon Tran
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyeonjeong Cho
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Minhee Kim
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Chaeyeong Kim
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - So Hee Kwon
- Department of Pharmacy, College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - SungJin Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jongsun Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Corresponding author. Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| | - Jisoo Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Corresponding author. Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
17
|
Daverey A, Agrawal SK. Pre and post treatment with curcumin and resveratrol protects astrocytes after oxidative stress. Brain Res 2018; 1692:45-55. [PMID: 29729252 DOI: 10.1016/j.brainres.2018.05.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 10/17/2022]
Abstract
The two most studied polyphenolic compounds, curcumin (Cur) and resveratrol (Res), have been reported to protect oxidative damage of astrocytes. The present study is designed to examine the comparative anti-oxidative effect of Cur and Res on astrocytes by studying their potential to protect H2O2 induced oxidative stress at 4 h and 24 h time exposure. The effect of Cur and Res on cell viability, ROS production, inflammation and astrogliosis was compared. The effect of these two on Nrf2 expression and its translocation to nuclear compartment was investigated. The results showed that both Cur and Res significantly increase astrocytes survival after oxidative stress at both time points, however, Res demonstrated better effect on cell viability than the Cur. Res, showing significant inhibition of ROS production at both time points. Cur displayed significant inhibition of ROS production at 4 h, suggesting that Cur is more active on ROS inhibition in the earlier phase of insult. Comparing the expression of NF-κB, Cur showed better anti-inflammatory action on NF-κB while Res did not have any effect of NF-κB expression at 4 h. Interestingly, Cur showed an upregulation of nuclear Nrf2 expression at 24 h whereas Res displayed no effect after 24 h incubation. Both Cur and Res inhibited the H2O2 induced translocation of Nrf2 into nucleus. In conclusion, based on our observation, we found that Cur and Res both protected astrocytes from oxidative stress. In addition, we observed that Cur is most effective in early hours of insult while Res is effective in late hours suggesting that Res may or may not have immediate effect on astrocytes.
Collapse
Affiliation(s)
- Amita Daverey
- Department of Surgery, Division of Neurosurgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sandeep K Agrawal
- Department of Surgery, Division of Neurosurgery, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
18
|
Abstract
The NFE2L2 gene encodes the transcription factor Nrf2 best known for regulating the expression of antioxidant and detoxification genes. Gene knockout approaches have demonstrated its universal cytoprotective features. While Nrf2 has been the topic of intensive research in cancer biology since its discovery in 1994, understanding the role of Nrf2 in cardiovascular disease has just begun. The literature concerning Nrf2 in experimental models of atherosclerosis, ischemia, reperfusion, cardiac hypertrophy, heart failure, and diabetes supports its cardiac protective character. In addition to antioxidant and detoxification genes, Nrf2 has been found to regulate genes participating in cell signaling, transcription, anabolic metabolism, autophagy, cell proliferation, extracellular matrix remodeling, and organ development, suggesting that Nrf2 governs damage resistance as well as wound repair and tissue remodeling. A long list of small molecules, most derived from natural products, have been characterized as Nrf2 inducers. These compounds disrupt Keap1-mediated Nrf2 ubquitination, thereby prohibiting proteasomal degradation and allowing Nrf2 protein to accumulate and translocate to the nucleus, where Nrf2 interacts with sMaf to bind to ARE in the promoter of genes. Recently alternative mechanisms driving Nrf2 protein increase have been revealed, including removal of Keap1 by autophagy due to p62/SQSTM1 binding, inhibition of βTrCP or Synoviolin/Hrd1-mediated ubiquitination of Nrf2, and de novo Nrf2 protein translation. We review here a large volume of literature reporting historical and recent discoveries about the function and regulation of Nrf2 gene. Multiple lines of evidence presented here support the potential of dialing up the Nrf2 pathway for cardiac protection in the clinic.
Collapse
Affiliation(s)
- Qin M Chen
- Department of Pharmacology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Anthony J Maltagliati
- Department of Pharmacology, College of Medicine, University of Arizona , Tucson, Arizona
| |
Collapse
|
19
|
Koleini N, Nickel BE, Wang J, Roveimiab Z, Fandrich RR, Kirshenbaum LA, Cattini PA, Kardami E. Fibroblast growth factor-2-mediated protection of cardiomyocytes from the toxic effects of doxorubicin requires the mTOR/Nrf-2/HO-1 pathway. Oncotarget 2017; 8:87415-87430. [PMID: 29152091 PMCID: PMC5675643 DOI: 10.18632/oncotarget.20558] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/04/2017] [Indexed: 12/09/2022] Open
Abstract
Background Cardiotoxic side effects impose limits to the use of anti-tumour chemotherapeutic drugs such as doxorubicin (Dox). There is a need for cardioprotective strategies to prevent the multiple deleterious effects of Dox. Here, we examined the ability of administered fibroblast growth factor-2 (FGF-2), a cardioprotective protein that is synthesized as high and low molecular weight (Hi-, Lo-FGF-2) isoforms, to prevent Dox-induced: oxidative stress; cell death; lysosome dysregulation; and inactivation of potent endogenous protective pathways, such as the anti-oxidant/detoxification nuclear factor erythroid-2-related factor (Nrf-2), heme oxygenase-1 (HO-1) axis. Methods and Results Brief pre-incubation of neonatal rat cardiomyocyte cultures with either Hi- or Lo-FGF-2 reduced the Dox-induced: oxidative stress; apoptotic/necrotic cell death; lysosomal dysregulation; decrease in active mammalian target of Rapamycin (mTOR). FGF-2 isoforms prevented the Dox-induced downregulation of Nrf-2, and promoted robust increases in the Nrf-2-downstream targets including the cardioprotective protein HO-1, and p62/SQSTM1, a multifunctional scaffold protein involved in autophagy. Chloroquine, an autophagic flux inhibitor, caused a further increase in p62/SQSTM1, indicating intact autophagic flux in the FGF-2-treated groups. A selective inhibitor for HO-1, Tin-Protoporphyrin, prevented the FGF-2 protection against cell death. The mTOR inhibitor Rapamycin prevented FGF-2 protection, and blocked the FGF-2 effects on Nrf-2, HO-1 and p62/SQSTM1. Conclusions In an acute setting Hi- or Lo-FGF-2 protect cardiomyocytes against multiple Dox-induced deleterious effects, by a mechanism dependent on preservation of mTOR activity, Nrf-2 levels, and the upregulation of HO-1. Preservation/activation of endogenous anti-oxidant/detoxification defences by FGF-2 is a desirable property in the setting of Dox-cardiotoxicity.
Collapse
Affiliation(s)
- Navid Koleini
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Manitoba, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Barbara E Nickel
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Jie Wang
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Zeinab Roveimiab
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Robert R Fandrich
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Manitoba, Canada.,Department of Human Anatomy and Cell Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lorrie A Kirshenbaum
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Manitoba, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter A Cattini
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Elissavet Kardami
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Manitoba, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Human Anatomy and Cell Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
20
|
Barančík M, Grešová L, Barteková M, Dovinová I. Nrf2 as a key player of redox regulation in cardiovascular diseases. Physiol Res 2017; 65 Suppl 1:S1-S10. [PMID: 27643930 DOI: 10.33549/physiolres.933403] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The oxidative stress plays an important role in the development of cardiovascular diseases (CVD). In CVD progression an aberrant redox regulation was observed. In this regulation levels of reactive oxygen species (ROS) play an important role in cellular signaling, where Nrf2 is the key regulator of redox homeostasis. Keap1-Nrf2-ARE system regulates a great set of detoxificant and antioxidant enzymes in cells after ROS and electrophiles exposure. In this review we focus on radical-generating systems in cardiovascular system as well as on Nrf2 as a target against oxidative stress and a key player of redox regulation in cardiovascular diseases. We also summarize the current knowledge about the role of Nrf2 in pathophysiology of several CVD (hypertension, cardiac hypertrophy, cardiomyopathies) as well as in cardioprotection against myocardial ischemia/ reperfusion injury.
Collapse
Affiliation(s)
- M Barančík
- Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovakia.
| | | | | | | |
Collapse
|
21
|
G-Quadruplex in the NRF2 mRNA 5' Untranslated Region Regulates De Novo NRF2 Protein Translation under Oxidative Stress. Mol Cell Biol 2016; 37:MCB.00122-16. [PMID: 27736771 DOI: 10.1128/mcb.00122-16] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 09/27/2016] [Indexed: 12/26/2022] Open
Abstract
Inhibition of protein synthesis serves as a general measure of cellular consequences of chemical stress. A few proteins are translated selectively and influence cell fate. How these proteins can bypass the general control of translation remains unknown. We found that low to mild doses of oxidants induce de novo translation of the NRF2 protein. Here we demonstrate the presence of a G-quadruplex structure in the 5' untranslated region (UTR) of NRF2 mRNA, as measured by circular dichroism, nuclear magnetic resonance, and dimethylsulfate footprinting analyses. Such a structure is important for 5'-UTR activity, since its removal by sequence mutation eliminated H2O2-induced activation of the NRF2 5' UTR. Liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based proteomics revealed elongation factor 1 alpha (EF1a) as a protein binding to the G-quadruplex sequence. Cells responded to H2O2 treatment by increasing the EF1a protein association with NRF2 mRNA, as measured by RNA-protein interaction assays. The EF1a interaction with small and large subunits of ribosomes did not appear to change due to H2O2 treatment, nor did posttranslational modifications, as measured by two-dimensional (2-D) Western blot analysis. Since NRF2 encodes a transcription factor essential for protection against tissue injury, our data have revealed a novel mechanism of cellular defense involving de novo NRF2 protein translation governed by the EF1a interaction with the G-quadruplex in the NRF2 5' UTR during oxidative stress.
Collapse
|
22
|
Sun G, Li Y, Ji Z. Atorvastatin attenuates inflammation and oxidative stress induced by ischemia/reperfusion in rat heart via the Nrf2 transcription factor. Int J Clin Exp Med 2015; 8:14837-14845. [PMID: 26628965 PMCID: PMC4658854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/13/2015] [Indexed: 06/05/2023]
Abstract
The role of atorvastatin in inflammation and oxidative stress induced by ischemia/reperfusion is currently not well understood. The aim of this study was toinvestigate whether atorvastatin modulates neutrophil accumulation, TNF-α induction and oxidative stress and to examine the possible role of the nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway in an ischemia/reperfusion injured rat heart model. Rats were randomly assigned into tosham operation group, myocardial ischemia/reperfusion (MI/R) group, MI/R + atorvastatin group. Myocardial infarct area, myeloperoxidase (MPO), serum creatinine kinase (CK) and lactate dehydrogenase (LDH) levels were monitored. The results indicate that compared to MI/R, atorvastatin reduced myocardial infarction area, MPO level, serum CK and LDH levels, and both serum and myocardial TNF-αproduction. In addition, atorvastatin increased SOD and GSH-PX activity and decreased MDA content. Atorvastatin also enhanced levels of Nrf2 and heme oxygenase-1. In summary, our data suggests that atorvastatin exerts significant cardioprotective effects following myocardial ischemia, possibly through the activation of the Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Guoqiang Sun
- Department of Emergency, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Yubo Li
- Changchun Medical CollegeChangchun, Jilin, China
| | - Zhiyong Ji
- Department of Emergency, The First Hospital of Jilin UniversityChangchun, Jilin, China
| |
Collapse
|
23
|
Strom J, Xu B, Tian X, Chen QM. Nrf2 protects mitochondrial decay by oxidative stress. FASEB J 2015; 30:66-80. [PMID: 26340923 DOI: 10.1096/fj.14-268904] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 08/13/2015] [Indexed: 12/22/2022]
Abstract
Sublethal levels of oxidative stress are commonly associated with various pathophysiological conditions. Cardiomyocytes have the highest content of mitochondria among all cell types, allowing the study of mitochondria in cells surviving oxidative stress and address whether nuclear factor-erythroid-derived 2-related factor 2 (Nrf2) can reverse these changes. Mitochondria normally exist in elaborated networks, which were replaced by predominately individual punctuate mitochondria 24 h after exposure to a nonlethal dose of H2O2. Electron microscopy revealed that cells surviving H2O2 show swelling of mitochondria with disorganized cristae and areas of condensation. Measurements of functional mitochondria showed a H2O2 dose-dependent decrease over a course of 5 d. At the protein and mRNA levels, cells surviving H2O2 treatment show a reduction of mitochondrial components, cytochrome c, and cytochrome b. Nrf2 overexpression prevented H2O2 from inducing mitochondria morphologic changes and reduction of cytochrome b/c. Although Nrf2 is known as a transcription factor regulating antioxidant and detoxification genes, Nrf2 overexpression did not significantly reduce the level of protein oxidation. Instead, Nrf2 was found to associate with the outer mitochondrial membrane. Mitochondria prepared from the myocardium of Nrf2 knockout mice are more sensitive to permeability transition. Our data suggest that Nrf2 protects mitochondria from oxidant injury likely through direct interaction with mitochondria.
Collapse
Affiliation(s)
- Joshua Strom
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Beibei Xu
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Xiuqing Tian
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Qin M Chen
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
24
|
Cheng L, Jin Z, Zhao R, Ren K, Deng C, Yu S. Resveratrol attenuates inflammation and oxidative stress induced by myocardial ischemia-reperfusion injury: role of Nrf2/ARE pathway. Int J Clin Exp Med 2015; 8:10420-10428. [PMID: 26379832 PMCID: PMC4565215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/03/2015] [Indexed: 06/05/2023]
Abstract
The protective role of resveratrol in myocardial ischemia/reperfusion is not well understood. The aim of this study was to investigate whether resveratrol modulates inflammation and oxidative stress and the possible role of nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway in an ischemia/reperfusion injured rat heart model. Rats were randomly exposed to sham operation, myocardial ischemia/reperfusion (MI/R) alone, and MI/R + resveratrol. The results demonstrated that compared to MI/R, resveratrol improved cardiac function, reduced myocardial infarction area, myocardial myeloperoxidase (MPO) levels, serum creatinine kinase (CK) and lactate dehydrogenase (LDH) levels. Resveratrol also markedly enhanced the activities of antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX), and reduced the level of malondialdehyde (MDA) in MI/R rats. Resveratrol also enhanced levels of Nrf2 and heme oxygenase-1. In summary, these results demonstrated that resveratrol exerted significant antioxidant and cardioprotective effects following myocardial ischemia, possibly through the activation of the Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Liang Cheng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Zhenxiao Jin
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Rong Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Kai Ren
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Chao Deng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| | - Shiqiang Yu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University Xi'an, China
| |
Collapse
|
25
|
Aboonabi A, Singh I. Chemopreventive role of anthocyanins in atherosclerosis via activation of Nrf2-ARE as an indicator and modulator of redox. Biomed Pharmacother 2015; 72:30-6. [PMID: 26054672 DOI: 10.1016/j.biopha.2015.03.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/30/2015] [Indexed: 12/21/2022] Open
Abstract
Anthocyanins have been reported to induce the expression of enzymes involved in both cellular antioxidant defenses and attenuating inflammation-associated pathogenesis. Induction of such enzymes by edible anthocyanin largely accounts for their atherosclerosis chemo-protective activities. Nuclear factor erythroid 2-related factor 2 (Nrf2) plays an essential role in the coordinated induction of those genes encoding redox-responsive and cellular defense antioxidant enzyme termed antioxidant response element (ARE). Current studies have revealed that Nrf2-ARE signaling is involved in attenuating inflammation-associated pathogenesis such as atherosclerosis. Conversely, reduction in Nrf2 signaling leads to enhanced susceptibility to oxidative stress and inflammatory tissue injuries. The activation of Nrf2-ARE might inhibit the production of pro-inflammatory mediator including cyclooxygenase-2, chemokines, cytokines, cell adhesion molecules, and induction nitric oxide synthase. This review highlights the gene expression induced by dietary anthocyanin via Nrf2 signaling on redox-regulated transcription factor in atherosclerosis disorders.
Collapse
Affiliation(s)
- Anahita Aboonabi
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia.
| | - Indu Singh
- Heart Foundation Research Centre, Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia.
| |
Collapse
|
26
|
Richardson C, Yan S, Vestal CG. Oxidative stress, bone marrow failure, and genome instability in hematopoietic stem cells. Int J Mol Sci 2015; 16:2366-85. [PMID: 25622253 PMCID: PMC4346841 DOI: 10.3390/ijms16022366] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/06/2015] [Accepted: 01/16/2015] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen species (ROS) can be generated by defective endogenous reduction of oxygen by cellular enzymes or in the mitochondrial respiratory pathway, as well as by exogenous exposure to UV or environmental damaging agents. Regulation of intracellular ROS levels is critical since increases above normal concentrations lead to oxidative stress and DNA damage. A growing body of evidence indicates that the inability to regulate high levels of ROS leading to alteration of cellular homeostasis or defective repair of ROS-induced damage lies at the root of diseases characterized by both neurodegeneration and bone marrow failure as well as cancer. That these diseases may be reflective of the dynamic ability of cells to respond to ROS through developmental stages and aging lies in the similarities between phenotypes at the cellular level. This review summarizes work linking the ability to regulate intracellular ROS to the hematopoietic stem cell phenotype, aging, and disease.
Collapse
Affiliation(s)
- Christine Richardson
- Department of Biological Sciences, UNC Charlotte, 9201 University City Blvd., Woodward Hall Room 386B, Charlotte, NC 28223, USA.
| | - Shan Yan
- Department of Biological Sciences, UNC Charlotte, 9201 University City Blvd., Woodward Hall Room 386B, Charlotte, NC 28223, USA.
| | - C Greer Vestal
- Department of Biological Sciences, UNC Charlotte, 9201 University City Blvd., Woodward Hall Room 386B, Charlotte, NC 28223, USA.
| |
Collapse
|
27
|
Granados-Principal S, El-azem N, Pamplona R, Ramirez-Tortosa C, Pulido-Moran M, Vera-Ramirez L, Quiles JL, Sanchez-Rovira P, Naudí A, Portero-Otin M, Perez-Lopez P, Ramirez-Tortosa MC. Hydroxytyrosol ameliorates oxidative stress and mitochondrial dysfunction in doxorubicin-induced cardiotoxicity in rats with breast cancer. Biochem Pharmacol 2014; 90:25-33. [DOI: 10.1016/j.bcp.2014.04.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 04/03/2014] [Accepted: 04/03/2014] [Indexed: 01/22/2023]
|
28
|
Wang W, Li S, Wang H, Li B, Shao L, Lai Y, Horvath G, Wang Q, Yamamoto M, Janicki JS, Wang XL, Tang D, Cui T. Nrf2 enhances myocardial clearance of toxic ubiquitinated proteins. J Mol Cell Cardiol 2014; 72:305-15. [PMID: 24747945 DOI: 10.1016/j.yjmcc.2014.04.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 04/03/2014] [Accepted: 04/09/2014] [Indexed: 11/29/2022]
Abstract
Nuclear factor erythroid-2 related factor 2 (Nrf2) is a master transcription factor that controls the basal and inducible expression of a battery of antioxidant genes and other cytoprotective phase II detoxifying enzymes. While knockout of Nrf2 exaggerates cardiac pathological remodeling and dysfunction in diverse pathological settings, pharmacological activation of Nrf2 protects against cardiomyocyte injury and cardiac dysfunction. In contrast, there is also a concern that the chronic activation of Nrf2 secondary to oxidative stress is a contributing mechanism for the reductive stress-mediated heart failure. However, a direct link between cardiac specific activation of Nrf2 and cardiac protection or dysfunction in vivo remains to be established. Therefore, we investigated the effect of cardiomyocyte-specific transgenic activation of Nrf2 (Nrf2(ctg)) on cardiac pathological remodeling and dysfunction. We found that the cardiomyocyte-specific activation of Nrf2 suppressed myocardial oxidative stress as well as cardiac apoptosis, fibrosis, hypertrophy, and dysfunction in a setting of sustained pressure overload induced by transverse aortic arch constriction (TAC) in mice. Notably, the constitutive activation of Nrf2 increased the steady level of autophagosomes while decreasing the ubiquitinated protein aggregates in the heart after TAC. Nrf2 gene gain- and loss-of-function approaches revealed that Nrf2 enhances autophagosome formation and autophagic flux in cardiomyocytes. Unexpectedly, while Nrf2 minimally regulated apoptosis, it suppressed significantly the proteotoxic necrosis in cardiomyocytes. In addition, Nrf2 attenuated the proteocytotoxicity presumably via enhancing autophagy-mediated clearance of ubiquitinated protein aggregates in cardiomyocytes. Taken together, we demonstrated for the first time that cardiac specific activation of Nrf2 suppresses cardiac maladaptive remodeling and dysfunction most likely by enhancing autophagic clearance of toxic protein aggregates in the heart.
Collapse
Affiliation(s)
- Wenjuan Wang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan 250012, China; Department of Cell Biology and Anatomy, University of South Carolina, SC 29208, USA
| | - Siying Li
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan 250012, China; Department of Cell Biology and Anatomy, University of South Carolina, SC 29208, USA
| | - Hui Wang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan 250012, China; Department of Cell Biology and Anatomy, University of South Carolina, SC 29208, USA
| | - Bin Li
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan 250012, China; Department of Cell Biology and Anatomy, University of South Carolina, SC 29208, USA
| | - Lei Shao
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan 250012, China; Department of Cell Biology and Anatomy, University of South Carolina, SC 29208, USA
| | - Yimu Lai
- Department of Cell Biology and Anatomy, University of South Carolina, SC 29208, USA
| | - Gary Horvath
- Department of Chemistry and Biochemistry, University of South Carolina, SC 29208, USA
| | - Qian Wang
- Department of Chemistry and Biochemistry, University of South Carolina, SC 29208, USA
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Joseph S Janicki
- Department of Cell Biology and Anatomy, University of South Carolina, SC 29208, USA
| | - Xing Li Wang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Dongqi Tang
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan 250012, China; Department of Cell Biology and Anatomy, University of South Carolina, SC 29208, USA.
| | - Taixing Cui
- Shandong University Qilu Hospital Research Center for Cell Therapy, Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan 250012, China; Department of Cell Biology and Anatomy, University of South Carolina, SC 29208, USA.
| |
Collapse
|
29
|
Zhou C, Li L, Li H, Gong J, Fang N. Delayed remote preconditioning induces cardioprotection: role of heme oxygenase-1. J Surg Res 2014; 191:51-7. [PMID: 24746951 DOI: 10.1016/j.jss.2014.03.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 01/25/2014] [Accepted: 03/18/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND The role of heme oxygenase-1 (HO-1) in the cardioprotection induced by delayed remote ischemic preconditioning (DRIPC) has not been investigated. Therefore, this study was designed to investigate whether HO-1 is involved in DRIPC-mediated cardioprotection in an isolated perfused rat heart model. MATERIALS AND METHODS Isolated rat hearts were subjected to 30 min ischemia followed by 60 min reperfusion. DRIPC (four cycles 5-min occlusion and 5-min reflow at the unilateral hind limb once per day for 1, 2, or 3 d before heart isolation, abbreviated as D1RIPC, D2RIPC, or D3RIPC respectively). Infarct size, myocardial troponin levels, and heart function were measured. The protein and messenger RNA levels of HO-1 were determined. RESULTS DRIPC facilitated postischemic cardiac functional recovery and decreased cardiac enzyme release. The infarct size-limiting effect of DRIPC was more pronounced in the D3RIPC group (10.22 ± 2.57%) than the D1RIPC group (22.34 ± 4.02%, P < 0.001) or the D2RIPC group (14.60 ± 3.13%, P = 0.034). These effects in the D1RIPC group could be blocked by Zinc Protoporphyrin IX (ZnPP) (an HO-1 specific inhibitor). DRIPC-mediated cardioprotection was associated with enhanced HO-1 protein expression (D1RIPC, 0.11 ± 0.03; versus 0.15 ± 0.06 in the D2RIPC group, P = 0.06; versus 0.20 ± 0.04 in the D3RIPC group, P = 0.04) and messenger RNA levels of HO-1 expression. CONCLUSIONS Our findings suggest that HO-1 is involved in the cardioprotection induced by DRIPC, and that increase in the number of preconditioning stimuli may enhance cardioprotective effects accompanied with increased HO-1 level.
Collapse
Affiliation(s)
- Chenghui Zhou
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing China
| | - Lihuan Li
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing China.
| | - Huatong Li
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing China
| | - Junsong Gong
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing China
| | - Nengxin Fang
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing China
| |
Collapse
|
30
|
Zhou C, Li H, Yao Y, Li L. Delayed remote ischemic preconditioning produces an additive cardioprotection to sevoflurane postconditioning through an enhanced heme oxygenase 1 level partly via nuclear factor erythroid 2-related factor 2 nuclear translocation. J Cardiovasc Pharmacol Ther 2014; 19:558-66. [PMID: 24651515 DOI: 10.1177/1074248414524479] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although both sevoflurane postconditioning (SPoC) and delayed remote ischemic preconditioning (DRIPC) have been proved effective in various animal and human studies, the combined effect of these 2 strategies remains unclear. Therefore, this study was designed to investigate this effect and elucidate the related signal mechanisms in a Langendorff perfused rat heart model. After 30-minute balanced perfusion, isolated hearts were subjected to 30-minute ischemia followed by 60-minute reperfusion except 90-minute perfusion for control. A synergic cardioprotective effect of SPoC (3% v/v) and DRIPC (4 cycles 5-minute occlusion/5-minute reflow at the unilateral hindlimb once per day for 3 days before heart isolation) was observed with facilitated cardiac functional recovery and decreased cardiac enzyme release. The infarct size-limiting effect was more pronounced in the combined group (6.76% ± 2.18%) than in the SPoC group (16.50% ± 4.55%, P < .001) or in the DRIPC group (10.22% ± 2.57%, P = .047). Subsequent analysis revealed that an enhanced heme oxygenase 1 (HO-1) expression, but not protein kinase B/AKt or extracellular signal-regulated kinase 1 and 2 activation, was involved in the synergic cardioprotective effect, which was further confirmed in the messenger RNA level of HO-1. Such trend was also observed in the nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, an upstream regulation of HO-1. In addition, correlation analysis showed a significantly positive relationship between HO-1 expression and Nrf2 translocation (r = 0.729, P < .001). Hence, we conclude that DRIPC may produce an additive cardioprotection to SPoC through an enhanced HO-1 expression partly via Nrf2 translocation.
Collapse
Affiliation(s)
- Chenghui Zhou
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huatong Li
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuntai Yao
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lihuan Li
- Department of Anesthesiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Sikalidis AK, Mazor KM, Lee JI, Roman HB, Hirschberger LL, Stipanuk MH. Upregulation of capacity for glutathione synthesis in response to amino acid deprivation: regulation of glutamate-cysteine ligase subunits. Amino Acids 2014; 46:1285-96. [PMID: 24557597 DOI: 10.1007/s00726-014-1687-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 01/29/2014] [Indexed: 12/22/2022]
Abstract
Using HepG2/C3A cells and MEFs, we investigated whether induction of GSH synthesis in response to sulfur amino acid deficiency is mediated by the decrease in cysteine levels or whether it requires a decrease in GSH levels per se. Both the glutamate-cysteine ligase catalytic (GCLC) and modifier (GCLM) subunit mRNA levels were upregulated in response to a lack of cysteine or other essential amino acids, independent of GSH levels. This upregulation did not occur in MEFs lacking GCN2 (general control non-derepressible 2, also known as eIF2α kinase 4) or in cells expressing mutant eIF2α lacking the eIF2α kinase Ser(51) phosphorylation site, indicating that expression of both GCLC and GCLM was mediated by the GCN2/ATF4 stress response pathway. Only the increase in GCLM mRNA level, however, was accompanied by a parallel increase in protein expression, suggesting that the enhanced capacity for GSH synthesis depended largely on increased association of GCLC with its regulatory subunit. Upregulation of both GCLC and GLCM mRNA levels in response to cysteine deprivation was dependent on new protein synthesis, which is consistent with expression of GCLC and GCLM being mediated by proteins whose synthesis depends on activation of the GCN2/ATF4 pathway. Our data suggest that the regulation of GCLC expression may be mediated by changes in the abundance of transcriptional regulators, whereas the regulation of GCLM expression may be mediated by changes in the abundance of mRNA stabilizing or destabilizing proteins. Upregulation of GCLM levels in response to low cysteine levels may serve to protect the cell in the face of a future stress requiring GSH as an antioxidant or conjugating/detoxifying agent.
Collapse
Affiliation(s)
- Angelos K Sikalidis
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA
| | | | | | | | | | | |
Collapse
|
32
|
Kumar H, Kim IS, More SV, Kim BW, Choi DK. Natural product-derived pharmacological modulators of Nrf2/ARE pathway for chronic diseases. Nat Prod Rep 2014; 31:109-39. [DOI: 10.1039/c3np70065h] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
33
|
Covas G, Marinho HS, Cyrne L, Antunes F. Activation of Nrf2 by H2O2: de novo synthesis versus nuclear translocation. Methods Enzymol 2013; 528:157-71. [PMID: 23849864 DOI: 10.1016/b978-0-12-405881-1.00009-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The most common mechanism described for the activation of the transcription factor Nrf2 is based on the inhibition of its degradation in the cytosol followed by its translocation to the nucleus. Recently, Nrf2 de novo synthesis was proposed as an additional mechanism for the rapid upregulation of Nrf2 by hydrogen peroxide (H2O2). Here, we describe a detailed protocol, including solutions, pilot experiments, and experimental setups, which allows exploring the role of H2O2, delivered either as a bolus or as a steady state, in endogenous Nrf2 translocation and synthesis. We also show experimental data, illustrating that H2O2 effects on Nrf2 activation in HeLa cells are strongly dependent both on the H2O2 concentration and on the method of H2O2 delivery. The de novo synthesis of Nrf2 is triggered within 5min of exposure to low concentrations of H2O2, preceding Nrf2 translocation to the nucleus which is slower. Evidence of de novo synthesis of Nrf2 is observed only for low H2O2 steady-state concentrations, a condition that is prevalent in vivo. This study illustrates the applicability of the steady-state delivery of H2O2 to uncover subtle regulatory effects elicited by H2O2 in narrow concentration and time ranges.
Collapse
Affiliation(s)
- Gonçalo Covas
- Departamento de Química e Bioquímica and Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | | | | | | |
Collapse
|
34
|
Fourtounis J, Wang IM, Mathieu MC, Claveau D, Loo T, Jackson AL, Peters MA, Therien AG, Boie Y, Crackower MA. Gene expression profiling following NRF2 and KEAP1 siRNA knockdown in human lung fibroblasts identifies CCL11/Eotaxin-1 as a novel NRF2 regulated gene. Respir Res 2012; 13:92. [PMID: 23061798 PMCID: PMC3546844 DOI: 10.1186/1465-9921-13-92] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/10/2012] [Indexed: 01/21/2023] Open
Abstract
Background Oxidative Stress contributes to the pathogenesis of many diseases. The NRF2/KEAP1 axis is a key transcriptional regulator of the anti-oxidant response in cells. Nrf2 knockout mice have implicated this pathway in regulating inflammatory airway diseases such as asthma and COPD. To better understand the role the NRF2 pathway has on respiratory disease we have taken a novel approach to define NRF2 dependent gene expression in a relevant lung system. Methods Normal human lung fibroblasts were transfected with siRNA specific for NRF2 or KEAP1. Gene expression changes were measured at 30 and 48 hours using a custom Affymetrix Gene array. Changes in Eotaxin-1 gene expression and protein secretion were further measured under various inflammatory conditions with siRNAs and pharmacological tools. Results An anti-correlated gene set (inversely regulated by NRF2 and KEAP1 RNAi) that reflects specific NRF2 regulated genes was identified. Gene annotations show that NRF2-mediated oxidative stress response is the most significantly regulated pathway, followed by heme metabolism, metabolism of xenobiotics by Cytochrome P450 and O-glycan biosynthesis. Unexpectedly the key eosinophil chemokine Eotaxin-1/CCL11 was found to be up-regulated when NRF2 was inhibited and down-regulated when KEAP1 was inhibited. This transcriptional regulation leads to modulation of Eotaxin-1 secretion from human lung fibroblasts under basal and inflammatory conditions, and is specific to Eotaxin-1 as NRF2 or KEAP1 knockdown had no effect on the secretion of a set of other chemokines and cytokines. Furthermore, the known NRF2 small molecule activators CDDO and Sulphoraphane can also dose dependently inhibit Eotaxin-1 release from human lung fibroblasts. Conclusions These data uncover a previously unknown role for NRF2 in regulating Eotaxin-1 expression and further the mechanistic understanding of this pathway in modulating inflammatory lung disease.
Collapse
Affiliation(s)
- Jimmy Fourtounis
- Department of Respiratory and Immunology, Merck Research Laboratories, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhang J, Dinh TN, Kappeler K, Tsaprailis G, Chen QM. La autoantigen mediates oxidant induced de novo Nrf2 protein translation. Mol Cell Proteomics 2012; 11:M111.015032. [PMID: 22207702 PMCID: PMC3433904 DOI: 10.1074/mcp.m111.015032] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 12/25/2011] [Indexed: 01/20/2023] Open
Abstract
Nrf2 gene encodes a transcription factor that regulates the expression of a cluster of antioxidant and detoxification genes. Recent works from our laboratory indicate that oxidative stress causes rapid de novo synthesis of Nrf2 protein. We have found that 5' Untranslated Region (5'UTR) of Nrf2 allows the mRNA to undergo an Internal Ribosomal Entry Site (IRES) mediated protein translation. Using liquid chromatography tandem MS, we have discovered that La/SSB protein bound to Nrf2 5'UTR in response to oxidative stress. In vitro RNA binding and in vivo ribonucleoprotein immunoprecipitation showed H(2)O(2) dose and time dependent increases of La/SSB binding to Nrf2 5'UTR. La/SSB protein translocated from the nuclei to cytoplasm and distributed in the perinuclear space in cells treated with H(2)O(2). Isolation of ribosomal fractions indicated that oxidants caused an association of La/SSB with ribosomes. Physical interaction of La/SSB with representative proteins from the small or large subunits of ribosomes was found to increase in cells responding to H(2)O(2) treatment. Knocking down La/SSB gene with siRNA prevented Nrf2 protein elevation or Nrf2 5'UTR activation by oxidants. In contrast, overexpression of La/SSB gene was able to enhance Nrf2 5'UTR activation and Nrf2 protein increase. Our data suggest that oxidants cause nuclear export of La/SSB protein and subsequent association of La/SSB with Nrf2 5'UTR and ribosomes. These events contribute to de novo Nrf2 protein translation because of oxidative stress.
Collapse
Affiliation(s)
- Jack Zhang
- From the ‡Department of Pharmacology, University of Arizona, College of Medicine, 1501 N. Campbell Ave, Tucson, Arizona 85724
| | - Thai Nho Dinh
- From the ‡Department of Pharmacology, University of Arizona, College of Medicine, 1501 N. Campbell Ave, Tucson, Arizona 85724
| | - Kyle Kappeler
- From the ‡Department of Pharmacology, University of Arizona, College of Medicine, 1501 N. Campbell Ave, Tucson, Arizona 85724
| | - George Tsaprailis
- §Center for Toxicology, College of Pharmacy, 1703 E. Mabel St Tucson, Arizona 85721
| | - Qin M. Chen
- From the ‡Department of Pharmacology, University of Arizona, College of Medicine, 1501 N. Campbell Ave, Tucson, Arizona 85724
| |
Collapse
|
36
|
Kappeler KV, Zhang J, Dinh TN, Strom JG, Chen QM. Histone deacetylase 6 associates with ribosomes and regulates de novo protein translation during arsenite stress. Toxicol Sci 2012; 127:246-55. [PMID: 22367689 DOI: 10.1093/toxsci/kfs070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) is known as a cytoplasmic enzyme that regulates cell migration, cell adhesion, and degradation of misfolded proteins by deacetylating substrates such as α-tubulin and Hsp90. When HaCaT keratinocytes were exposed to 1-200μM sodium arsenite, we observed perinuclear localization of HDAC6 within 30 min. Although the overall level of HDAC6 protein did not change, sodium arsenite caused an increase of HDAC6 in ribosomal fractions. Separation of ribosomal subunits versus intact ribosomes or polysomes indicated that HDAC6 was mainly detected in 40/43S fractions containing the small ribosomal subunit in untreated cells but was associated with 40/43S and 60/80S ribosomal fractions in arsenite-treated cells. Immunocytochemistry studies revealed that arsenite caused colocalization of HDAC6 with the ribosomal large and small subunit protein L36a and S6. Both L36a and S6 were detected in the immunocomplex of HDAC6 isolated from arsenite-treated cells. The observed physical interaction of HDAC6 with ribosomes pointed to a role of HDAC6 in stress-induced protein translation. Among arsenite stress-induced proteins, de novo Nrf2 protein translation was inhibited by Tubastatin A. These data demonstrate that HDAC6 was recruited to ribosomes, physically interacted with ribosomal proteins, and regulated de novo protein translation in keratinocytes responding to arsenite stress.
Collapse
Affiliation(s)
- Kyle V Kappeler
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724, USA
| | | | | | | | | |
Collapse
|
37
|
Synchrotron radiation induced X-ray emission studies of the antioxidant mechanism of the organoselenium drug ebselen. J Biol Inorg Chem 2012; 17:589-98. [DOI: 10.1007/s00775-012-0879-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 01/23/2012] [Indexed: 01/30/2023]
|
38
|
Angeloni C, Motori E, Fabbri D, Malaguti M, Leoncini E, Lorenzini A, Hrelia S. H2O2 preconditioning modulates phase II enzymes through p38 MAPK and PI3K/Akt activation. Am J Physiol Heart Circ Physiol 2011; 300:H2196-205. [PMID: 21478407 DOI: 10.1152/ajpheart.00934.2010] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ischemic preconditioning is a complex cardioprotective phenomenon that involves adaptive changes in cells and molecules and occurs in a biphasic pattern: an early phase after 1-2 h and a late phase after 12-24 h. While it is widely accepted that reactive oxygen species are strongly involved in triggering ischemic preconditiong, it is not clear if they play a major role in the early or late phase of preconditioning and which are the mechanisms involved. The present study was designed to investigate the mechanisms behind H(2)O(2)-induced cardioprotection in rat neonatal cardiomyocytes. We focused on antioxidant and phase II enzymes and their modulation by protein kinase signaling pathways and nuclear-factor-E(2)-related factor-1 (Nrf1) and Nrf2. H(2)O(2) preconditioning was able to counteract oxidative stress more effectively in the late than in the early phase of adaptation. In particular, H(2)O(2) preconditioning counteracted oxidative stress-induced apoptosis by decreasing caspase-3 activity, increasing Bcl2 expression and selectively increasing the expression and activity of antioxidant and phase II enzymes through Nrf1 and Nrf2 translocation to the nucleus. The downregulation of Nrf1 and Nrf2 by small interfering RNA reduced the expression level of phase II enzymes. Specific inhibitors of phosphatidylinositol 3-kinase/Akt and p38 MAPK activation partially reduced the cardioprotection elicited by H(2)O(2) preconditioning and the induction and activity of phase II enzymes. These findings demonstrate, for the first time, a key role for Nrf1, and not only for Nrf2, in the induction of phase II enzymes triggered by H(2)O(2) preconditioning.
Collapse
Affiliation(s)
- Cristina Angeloni
- Università di Bologna, Dipartimento di Biochimica "G. Moruzzi," Via Irnerio 48, 40126 Bologna, Italy.
| | | | | | | | | | | | | |
Collapse
|
39
|
MOUZANNAR R, MCCAFFERTY J, BENEDETTO G, RICHARDSON C. TRANSCRIPTIONAL AND PHOSPHO-PROTEOMIC SCREENS REVEAL STEM CELL ACTIVATION OF INSULIN-RESISTANCE AND TRANSFORMATION PATHWAYS FOLLOWING A SINGLE MINIMALLY TOXIC EPISODE OF ROS. INTERNATIONAL JOURNAL OF GENOMICS AND PROTEOMICS 2011; 2:34-49. [PMID: 21743783 PMCID: PMC3131088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Elevated reactive oxidative species (ROS) are cytotoxic, and chronic elevated levels of ROS have been implicated in multiple diseases as well as cellular transformation and tumor progression. However, the potential for a transient and minimally toxic episode of ROS exposure, or a minimal threshold dose of ROS, to initiate disease or cellular transformation is unclear. We examined both transcriptional and phospho-proteomic responses of murine embryonic stem (ES) cells to a single brief exposure of minimally toxic hydrogen peroxide (H(2)O(2)). The cellular response was distinct from those induced by either an acute exposure to H(2)O(2) or the topoisomerase II poison etoposide. Analysis of tumorigenesis-related transcripts revealed a significant up-regulation of oncogenes and down-regulation of tumor suppressors. Analysis of the phospho-proteomic response demonstrated insulin-signaling induction, including insulin receptor Y972 hypophosphorylation, similar to insulin-resistance mouse models and observed in diabetic patients. In addition, ES cells were more resistant to ROS than differentiated cells, and retained their transcriptional self-renewal signature, suggesting stem cells have a higher potential for ROS-mediated mutagenesis and proliferation in vivo. These results are a direct demonstration that even brief and non-toxic exposures to ROS may induce transduction of insulin resistance and transformation signaling in stem cells leading to diabetes and cancer.
Collapse
Affiliation(s)
- R. MOUZANNAR
- UNC-Charlotte, Department of Biology and Bioinformatics Research Center, Charlotte, NC 28223
| | - J. MCCAFFERTY
- UNC-Charlotte, Department of Bioinformatics and Genomics, Charlotte, NC 28223
| | - G. BENEDETTO
- UNC-Charlotte, Department of Biology and Bioinformatics Research Center, Charlotte, NC 28223
| | - C. RICHARDSON
- UNC-Charlotte, Department of Biology and Bioinformatics Research Center, Charlotte, NC 28223
| |
Collapse
|
40
|
Chen N, Ko M. Schisandrin B-induced glutathione antioxidant response and cardioprotection are mediated by reactive oxidant species production in rat hearts. Biol Pharm Bull 2010; 33:825-9. [PMID: 20460761 DOI: 10.1248/bpb.33.825] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To investigate the involvement of reactive oxidant species (ROS), presumably arising from cytochrome P-450 (CYP)-catalyzed metabolism of schisandrin B (Sch B), in triggering glutathione antioxidant response, Sch B induced reduced nicotinamide adenine dinucleotide phosphate (NADPH)-dependent and CYP-catalyzed reaction and associated ROS production were examined in rat heart microsomes. Sch B analogs were also studied for comparison. Using rat heart microsomes as a source of CYP, Sch B and schisandrin C (Sch C), but not schisandrin A and dimethyl diphenyl bicarboxylate (an intermediate compound derived from the synthesis of Sch C), were found to serve as co-substrate for the CYP-catalyzed NADPH oxidation reaction, with concomitant production of ROS. The stimulation of CYP-catalyzed NADPH oxidation reaction and/or ROS production by Sch B or Sch C correlated with the increase in mitochondrial reduced glutathione level and protection against ischemia/reperfusion (I/R) injury in rat hearts. The involvement of ROS in Sch B-induced cardioprotection was further confirmed by the suppressive effect produced by N-acetylcysteine or alpha-tocopherol pretreatment. Taken together, these results suggest that Sch B-induced glutathione antioxidant response and cardioprotection may be mediated by ROS arising from CYP-catalyzed reaction.
Collapse
Affiliation(s)
- Na Chen
- Department of Biochemistry, Hong Kong University of Science & Technology, Clear Water Bay, Hong Kong SAR, China
| | | |
Collapse
|
41
|
Hong Y, Yan W, Chen S, Sun CR, Zhang JM. The role of Nrf2 signaling in the regulation of antioxidants and detoxifying enzymes after traumatic brain injury in rats and mice. Acta Pharmacol Sin 2010; 31:1421-30. [PMID: 20953205 DOI: 10.1038/aps.2010.101] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
AIM To determine whether Nrf2 signaling pathway activation could attenuate oxidative stress and neuronal damage following traumatic brain injury (TBI). METHODS Controlled cortical impact (CCI) injury was performed in Sprague-Dawley rats and Nrf2-knockout or control mice. Sulforaphane (SFN), a potent Nrf2 activator, was used to activate Nrf2. Oxidative stress, lesion volume, neuron degeneration, and neurologic dysfunction were determined using biochemical, histopathological and neuroethologic approaches. Protein and mRNA levels of Nrf2 and the antioxidant enzymes heme oxygenase 1 (HO-1) and NAD(P)H:quinine oxidoreductase 1 (NQO1) were assessed using Western blot analysis and RT-PCR. RESULTS Activation of Nrf2 by SFN( 5 mg/kg, ip) induced the nuclear translocation and activation of Nrf2, which resulted in an up-regulation of Nrf2-dependent antioxidant enzymes and a reduction of oxidative damage after TBI. In accordance with these biochemical changes, SFN also significantly reduced neuronal death, contusion volume, and neurological dysfunction after TBI. Furthermore, Nrf2-knockout mice showed more severe oxidative stress and neurologic deficits after TBI and did not benefit from the effects of SFN. CONCLUSION Nrf2 plays a pivotal role in cell defenses against the oxidative stress of TBI. In addition, pharmacological activation of the Nrf2 signaling pathway by small molecule inducers such as SFN attenuated oxidative stress and neuronal damage following TBI.
Collapse
|
42
|
Morrissy S, Xu B, Aguilar D, Zhang J, Chen QM. Inhibition of apoptosis by progesterone in cardiomyocytes. Aging Cell 2010; 9:799-809. [PMID: 20726854 DOI: 10.1111/j.1474-9726.2010.00619.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
While gender-based differences in heart disease have raised the possibility that estrogen (ES) or progesterone (PG) may have cardioprotective effects, recent controversy regarding hormone replacement therapy has questioned the cardiac effects of these steroids. Using cardiomyocytes, we tested whether ES or PG has protective effects at the cellular level. We found that PG but not ES protects cardiomyocytes from apoptotic cell death induced by doxorubicin (Dox). PG inhibited apoptosis in a dose-dependent manner, by 12 ± 4.0% at 1 μm and 60 ± 1.0% at 10 μm. The anti-apoptotic effect of PG was also time dependent, causing 18 ± 5% or 62 + 2% decrease in caspase-3 activity within 1 h or 72 h of pretreatment. While PG causes nuclear translocation of its receptor within 20 min, the cytoprotective effect of PG was canceled by mifepristone (MF), a PG receptor antagonist. Analyses using Affymetrix high-density oligonucleotide array and RT-PCR found that PG induced Bcl-xL, metallothionine, NADPH quinone oxidoreductase 1, glutathione peroxidase-3, and four isoforms of glutathione S-transferase. Western blot analyses revealed that PG indeed induced an elevation of Bcl-xL protein in a dose- and time-dependent manner. Nuclear run-on assay indicated that PG induced Bcl-xL gene transcription. Inhibiting the expression of Bcl-xL using siRNA reduced the cytoprotective effect of PG. Our data suggests that PG induces a cytoprotective effect in cardiomyocytes in association with induction of Bcl-xL gene.
Collapse
Affiliation(s)
- Stephen Morrissy
- Department of Pharmacology, University of Arizona, Tucson, 85724, USA
| | | | | | | | | |
Collapse
|
43
|
4-hydroxy-2-nonenal protects against cardiac ischemia-reperfusion injury via the Nrf2-dependent pathway. J Mol Cell Cardiol 2010; 49:576-86. [PMID: 20685357 DOI: 10.1016/j.yjmcc.2010.05.011] [Citation(s) in RCA: 297] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2009] [Revised: 04/24/2010] [Accepted: 05/21/2010] [Indexed: 11/24/2022]
Abstract
Reactive oxygen species (ROS) attack polyunsaturated fatty acids of the membrane and trigger lipid peroxidation, which results in the generation of alpha,beta-unsaturated aldehydes, such as 4-hydroxy-2-nonenal (4-HNE). There is compelling evidence that high concentrations of aldehydes are responsible for much of the damage elicited by cardiac ischemia-reperfusion injury, while sublethal concentrations of aldehydes stimulate stress resistance pathways, to achieve cardioprotection. We investigated the mechanism of cardioprotection mediated by 4-HNE. For cultured cardiomyocytes, 4-HNE was cytotoxic at higher concentrations (>or=20 microM) but had no appreciable cytotoxicity at lower concentrations. Notably, a sublethal concentration (5muM) of 4-HNE primed cardiomyocytes to become resistant to cytotoxic concentrations of 4-HNE. 4-HNE induced nuclear translocation of transcription factor NF-E2-related factor 2 (Nrf2), and enhanced the expression of gamma-glutamylcysteine ligase (GCL) and the core subunit of the Xc(-) high-affinity cystine transporter (xCT), thereby increasing 1.45-fold the intracellular GSH levels. Cardiomyocytes treated with either Nrf2-specific siRNA or the GCL inhibitor l-buthionine sulfoximine (BSO) were less tolerant to 4-HNE. Moreover, the cardioprotective effect of 4-HNE pretreatment against subsequent glucose-free anoxia followed by reoxygenation was completely abolished in these cells. Intravenous administration of 4-HNE (4 mg/kg) activated Nrf2 in the heart and increased the intramyocardial GSH content, and consequently improved the functional recovery of the left ventricle following ischemia-reperfusion in Langendorff-perfused hearts. This cardioprotective effect of 4-HNE was not observed for Nrf2-knockout mice. In summary, 4-HNE activates Nrf2-mediated gene expression and stimulates GSH biosynthesis, thereby conferring on cardiomyocytes protection against ischemia-reperfusion injury.
Collapse
|
44
|
Malhotra D, Portales-Casamar E, Singh A, Srivastava S, Arenillas D, Happel C, Shyr C, Wakabayashi N, Kensler TW, Wasserman WW, Biswal S. Global mapping of binding sites for Nrf2 identifies novel targets in cell survival response through ChIP-Seq profiling and network analysis. Nucleic Acids Res 2010; 38:5718-34. [PMID: 20460467 PMCID: PMC2943601 DOI: 10.1093/nar/gkq212] [Citation(s) in RCA: 613] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The Nrf2 (nuclear factor E2 p45-related factor 2) transcription factor responds to diverse oxidative and electrophilic environmental stresses by circumventing repression by Keap1, translocating to the nucleus, and activating cytoprotective genes. Nrf2 responses provide protection against chemical carcinogenesis, chronic inflammation, neurodegeneration, emphysema, asthma and sepsis in murine models. Nrf2 regulates the expression of a plethora of genes that detoxify oxidants and electrophiles and repair or remove damaged macromolecules, such as through proteasomal processing. However, many direct targets of Nrf2 remain undefined. Here, mouse embryonic fibroblasts (MEF) with either constitutive nuclear accumulation (Keap1−/−) or depletion (Nrf2−/−) of Nrf2 were utilized to perform chromatin-immunoprecipitation with parallel sequencing (ChIP-Seq) and global transcription profiling. This unique Nrf2 ChIP-Seq dataset is highly enriched for Nrf2-binding motifs. Integrating ChIP-Seq and microarray analyses, we identified 645 basal and 654 inducible direct targets of Nrf2, with 244 genes at the intersection. Modulated pathways in stress response and cell proliferation distinguish the inducible and basal programs. Results were confirmed in an in vivo stress model of cigarette smoke-exposed mice. This study reveals global circuitry of the Nrf2 stress response emphasizing Nrf2 as a central node in cell survival response.
Collapse
Affiliation(s)
- Deepti Malhotra
- Department of Environmental Health Sciences Department, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Curcumin Protects Rat Myocardium Against Isoproterenol-Induced Ischemic Injury: Attenuation of Ventricular Dysfunction Through Increased Expression of Hsp27 Alongwith Strengthening Antioxidant Defense System. J Cardiovasc Pharmacol 2010; 55:377-84. [DOI: 10.1097/fjc.0b013e3181d3da01] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Copple IM, Goldring CE, Kitteringham NR, Park BK. The keap1-nrf2 cellular defense pathway: mechanisms of regulation and role in protection against drug-induced toxicity. Handb Exp Pharmacol 2010:233-66. [PMID: 20020265 DOI: 10.1007/978-3-642-00663-0_9] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adverse drug reactions pose a significant public health problem. In some cases, the process of drug metabolism can contribute to the onset of toxicity through the bioactivation of a parent molecule to a chemically reactive intermediate. In order to maintain a favorable balance between bioactivation and detoxification, mammalian cells have evolved an inducible cell defense system known as the antioxidant response pathway. The activity of this cytoprotective pathway is largely regulated by the transcription factor Nrf2, which governs the expression of many phase II detoxification and antioxidant enzymes. In turn, the activity of Nrf2 is regulated by the cysteine-rich cytosolic inhibitor Keap1, which acts as a "sensor" for chemical/oxidative stress. This article summarizes our current understanding of the molecular mechanisms that regulate the function of the Keap1-Nrf2 pathway and highlights the importance of Nrf2 in the protection against drug-induced toxicity.
Collapse
Affiliation(s)
- Ian M Copple
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutics, The University of Liverpool, Sherrington Building, Ashton Street, Liverpool, Merseyside L69 3GE, UK
| | | | | | | |
Collapse
|
47
|
Kim J, Cha YN, Surh YJ. A protective role of nuclear factor-erythroid 2-related factor-2 (Nrf2) in inflammatory disorders. Mutat Res 2009; 690:12-23. [PMID: 19799917 DOI: 10.1016/j.mrfmmm.2009.09.007] [Citation(s) in RCA: 536] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 09/20/2009] [Accepted: 09/22/2009] [Indexed: 02/06/2023]
Abstract
Nuclear factor-erythroid 2-related factor-2 (Nrf2) is a key transcription factor that plays a central role in cellular defense against oxidative and electrophilic insults by timely induction of antioxidative and phase-2 detoxifying enzymes and related stress-response proteins. The 5'-flanking regions of genes encoding these cytoprotective proteins contain a specific consensus sequence termed antioxidant response element (ARE) to which Nrf2 binds. Recent studies have demonstrated that Nrf2-ARE signaling is also involved in attenuating inflammation-associated pathogenesis, such as autoimmune diseases, rheumatoid arthritis, asthma, emphysema, gastritis, colitis and atherosclerosis. Thus, disruption or loss of Nrf2 signaling causes enhanced susceptibility not only to oxidative and electrophilic stresses but also to inflammatory tissue injuries. During the early-phase of inflammation-mediated tissue damage, activation of Nrf2-ARE might inhibit the production or expression of pro-inflammatory mediators including cytokines, chemokines, cell adhesion molecules, matrix metalloproteinases, cyclooxygenase-2 and inducible nitric oxide synthase. It is likely that the cytoprotective function of genes targeted by Nrf2 may cooperatively regulate the innate immune response and also repress the induction of pro-inflammatory genes. This review highlights the protective role of Nrf2 in inflammation-mediated disorders with special focus on the inflammatory signaling modulated by this redox-regulated transcription factor.
Collapse
Affiliation(s)
- Jiyoung Kim
- National Research Laboratory, College of Pharmacy, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, Republic of Korea
| | | | | |
Collapse
|
48
|
Du Y, Villeneuve NF, Wang XJ, Sun Z, Chen W, Li J, Lou H, Wong PK, Zhang DD. Oridonin confers protection against arsenic-induced toxicity through activation of the Nrf2-mediated defensive response. ENVIRONMENTAL HEALTH PERSPECTIVES 2008; 116:1154-61. [PMID: 18795156 PMCID: PMC2535615 DOI: 10.1289/ehp.11464] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2008] [Accepted: 05/21/2008] [Indexed: 05/12/2023]
Abstract
BACKGROUND Groundwater contaminated with arsenic imposes a big challenge to human health worldwide. Using natural compounds to subvert the detrimental effects of arsenic represents an attractive strategy. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a critical regulator of the cellular antioxidant response and xenobiotic metabolism. Recently, activation of the Nrf2 signaling pathway has been reported to confer protection against arsenic-induced toxicity in a cell culture model. OBJECTIVES The goal of the present work was to identify a potent Nrf2 activator from plants as a chemopreventive compound and to demonstrate the efficacy of the compound in battling arsenic-induced toxicity. RESULTS Oridonin activated the Nrf2 signaling pathway at a low subtoxic dose and was able to stabilize Nrf2 by blocking Nrf2 ubiquitination and degradation, leading to accumulation of the Nrf2 protein and activation of the Nrf2-dependent cytoprotective response. Pretreatment of UROtsa cells with 1.4 muM oridonin significantly enhanced the cellular redox capacity, reduced formation of reactive oxygen species (ROS), and improved cell survival after arsenic challenge. CONCLUSIONS We identified oridonin as representing a novel class of Nrf2 activators and illustrated the mechanism by which the Nrf2 pathway is activated. Furthermore, we demonstrated the feasibility of using natural compounds targeting Nrf2 as a therapeutic approach to protect humans from various environmental insults that may occur daily.
Collapse
Affiliation(s)
- Yu Du
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
- Department of Natural Products, School of Pharmaceutical Sciences, Shandong University, Shandong, People’s Republic of China
| | - Nicole F. Villeneuve
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Xiao-Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Zheng Sun
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Weimin Chen
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Jixue Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Hongxiang Lou
- Department of Natural Products, School of Pharmaceutical Sciences, Shandong University, Shandong, People’s Republic of China
| | - Pak Kin Wong
- Aerospace and Mechanical Engineering Department, University of Arizona, Tucson, Arizona, USA
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
- Address correspondence to D.D. Zhang, University of Arizona, College of Pharmacy, 1703 East Mabel, Tucson, AZ 85721 USA. Telephone: (520) 626-9918. Fax: (520) 626-2466. E-mail:
| |
Collapse
|
49
|
Genomic and proteomic profiling of oxidative stress response in human diploid fibroblasts. Biogerontology 2008; 10:125-51. [PMID: 18654835 DOI: 10.1007/s10522-008-9157-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Accepted: 06/16/2008] [Indexed: 12/15/2022]
Abstract
A number of lines of evidence suggest that senescence of normal human diploid fibroblasts (HDFs) in culture is relevant to the process of aging in vivo. Using normal human skin diploid fibroblasts, we examine the changes in genes and proteins following treatment with a mild dose of H2O2, which induces premature senescence. Multidimensional Protein Identification Technology (MudPIT) in combination with mass spectrometry analyses of whole cell lysates from HDFs detected 65 proteins in control group, 48 proteins in H2O2-treated cells and 109 proteins common in both groups. In contrast, cDNA microarray analyses show 173 genes up-regulated and 179 genes down-regulated upon H2O2 treatment. Both MudPIT and cDNA microarray analyses indicate that H2O2 treatment caused elevated levels of thioredoxin reductase 1. Semi-quantitative RT-PCR and Western-blot were able to verify the finding. Out of a large number of genes or proteins detected, only a small fraction shows the overlap between the outcomes of microarray versus proteomics. The low overlap suggests the importance of considering proteins instead of transcripts when investigating the gene expression profile altered by oxidative stress.
Collapse
|
50
|
Ding Y, Choi KJ, Kim JH, Han X, Piao Y, Jeong JH, Choe W, Kang I, Ha J, Forman HJ, Lee J, Yoon KS, Kim SS. Endogenous hydrogen peroxide regulates glutathione redox via nuclear factor erythroid 2-related factor 2 downstream of phosphatidylinositol 3-kinase during muscle differentiation. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1529-41. [PMID: 18458092 DOI: 10.2353/ajpath.2008.070429] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We reported previously that endogenous reactive oxygen species (ROS) function as myogenic signaling molecules. It has also been determined that excess ROS induce electrophile-response element (EpRE)-driven gene expression via activation of nuclear factor erythroid 2-related factor 2 (Nrf2). Nonetheless, the relationship between the metabolism of ROS (eg, H(2)O(2)) through glutathione (GSH) up-regulation, GSH-dependent reduction of H(2)O(2), and Nrf2-dependent gene regulation is not well established. Therefore, we attempted to determine whether H(2)O(2) controls the intracellular GSH redox state via the Nrf2-glutamate-cysteine ligase (GCL)/glutathione reductase (GR)-GSH signaling pathway. In our experiments, enhanced H(2)O(2) generation was accompanied by an increase in both total GSH levels and the GSH/GSSG ratio during muscle differentiation. Both GCL and GR transcriptional expression levels were markedly increased during muscle differentiation but reduced by catalase treatment. Nrf2 protein expression and nuclear translocation increased during myogenesis. The inhibition of GCL, GR, and Nrf2 both by inhibitors and by RNA interference blocked muscle differentiation. Phosphatidylinositol 3-kinase regulated the expression of the GCL C (a catalytic subunit) and GR genes via the induction of Nrf2 nuclear translocation and expression. In conclusion, endogenous H(2)O(2) generated during muscle differentiation not only functions as a signaling molecule, but also regulates the GSH redox state via activation of the Nrf2-GCL/GR-GSH signaling pathway downstream of phosphatidylinositol 3-kinase.
Collapse
Affiliation(s)
- Yan Ding
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|