51
|
Zheng D, Liu Z, Zhou Y, Hou N, Yan W, Qin Y, Ye Q, Cheng X, Xiao Q, Bao Y, Luo J, Wu X. Urolithin B, a gut microbiota metabolite, protects against myocardial ischemia/reperfusion injury via p62/Keap1/Nrf2 signaling pathway. Pharmacol Res 2020; 153:104655. [PMID: 31996327 DOI: 10.1016/j.phrs.2020.104655] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 02/07/2023]
Abstract
Ischemia/reperfusion (IR) induces additional damage during the restoration of blood flow to ischemic myocardium. Urolithin B (UB) is one of the gut metabolites of ellagitannins, a class of antioxidant polyphenols, which was found to be protective against oxidative stress in multiple organs. However, the role of UB in cardiovascular disease remains elusive. Adult Sprague Dawley rats were subjected to left anterior descending artery ligation for 30 min followed by 120 min of reperfusion, with or without UB treatment. In vitro, the H9c2 cardiomyocytes were subjected to hypoxia (94 %N2/5 %CO2/1 %O2) for 3 h, followed by reoxygenation (74 %N2/5 %CO2/21 %O2) for 3 h (HR). UB was found to decrease myocardial infarct size and attenuate the cardiac dysfunction in the rats after IR, and protect against HR injury in H9c2 cardiomyocytes. Mechanistically, UB inhibited autophagy by activating Akt/mTOR/ULK1 pathway and protected against oxidative stress and caspase 3-dependent cell apoptosis. In particular, UB induced accumulation of p62 and its interaction with Keap1, which promoted Nrf2 nuclear translocation during HR insult. Of note, the protection of UB against superoxide production and apoptotic cell death was compromised with Nrf2 gene silencing. Taken together, our findings suggested that UB protected against myocardial IR injury at least partially via the p62/Keap1/Nrf2 signaling pathway, which highlights the potential of UB as a novel therapy for ischemic heart disease.
Collapse
Affiliation(s)
- Dechong Zheng
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, PR China
| | - Zumei Liu
- Department of Laboratory Medicine and Central Laboratories, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, PR China
| | - You Zhou
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China
| | - Ning Hou
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China
| | - Wei Yan
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Yuan Qin
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China
| | - Qianfang Ye
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China
| | - XinYi Cheng
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China
| | - Qing Xiao
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China
| | - Yonglin Bao
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China
| | - Jiandong Luo
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China; Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China.
| | - Xiaoqian Wu
- Key Laboratory of Molecular Clinical Pharmacology & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China; Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China.
| |
Collapse
|
52
|
Cellular microdomains for nitric oxide signaling in endothelium and red blood cells. Nitric Oxide 2020; 96:44-53. [PMID: 31911123 DOI: 10.1016/j.niox.2020.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/23/2019] [Accepted: 01/02/2020] [Indexed: 12/13/2022]
Abstract
There is accumulating evidence that biological membranes are not just homogenous lipid structures, but are highly organized in microdomains, i.e. compartmentalized areas of protein and lipid complexes, which facilitate necessary interactions for various signaling pathways. Each microdomain exhibits unique composition, membrane location and dynamics, which ultimately shape their functional characteristics. In the vasculature, microdomains are crucial for organizing and compartmentalizing vasodilatory signals that contribute to blood pressure homeostasis. In this review we aim to describe how membrane microdomains in both the endothelium and red blood cells allow context-specific regulation of the vasodilatory signal nitric oxide (NO) and its corresponding metabolic products, and how this results in tightly controlled systemic physiological responses. We will describe (1) structural characteristics of microdomains including lipid rafts and caveolae; (2) endothelial cell caveolae and how they participate in mechanosensing and NO-dependent mechanotransduction; (3) the myoendothelial junction of resistance arterial endothelial cells and how protein-protein interactions within it have profound systemic effects on blood pressure regulation, and (4) putative/proposed NO microdomains in RBCs and how they participate in control of systemic NO bioavailability. The sum of these discussions will provide a current view of NO regulation by cellular microdomains.
Collapse
|
53
|
Myricetin Alleviates Pathological Cardiac Hypertrophy via TRAF6/TAK1/MAPK and Nrf2 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6304058. [PMID: 31885808 PMCID: PMC6925812 DOI: 10.1155/2019/6304058] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/25/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
Myricetin (Myr) is a common plant-derived polyphenol and is well recognized for its multiple activities including antioxidant, anti-inflammation, anticancer, and antidiabetes. Our previous studies indicated that Myr protected mouse heart from lipopolysaccharide and streptozocin-induced injuries. However, it remained to be unclear whether Myr could prevent mouse heart from pressure overload-induced pathological hypertrophy. Wild type (WT) and cardiac Nrf2 knockdown (Nrf2-KD) mice were subjected to aortic banding (AB) surgery and then administered with Myr (200 mg/kg/d) for 6 weeks. Myr significantly alleviated AB-induced cardiac hypertrophy, fibrosis, and cardiac dysfunction in both WT and Nrf2-KD mice. Myr also inhibited phenylephrine- (PE-) induced neonatal rat cardiomyocyte (NRCM) hypertrophy and hypertrophic markers' expression in vitro. Mechanically, Myr markedly increased Nrf2 activity, decreased NF-κB activity, and inhibited TAK1/p38/JNK1/2 MAPK signaling in WT mouse hearts. We further demonstrated that Myr could inhibit TAK1/p38/JNK1/2 signaling via inhibiting Traf6 ubiquitination and its interaction with TAK1 after Nrf2 knockdown in NRCM. These results strongly suggested that Myr could attenuate pressure overload-induced pathological hypertrophy in vivo and PE-induced NRCM hypertrophy via enhancing Nrf2 activity and inhibiting TAK1/P38/JNK1/2 phosphorylation by regulating Traf6 ubiquitination. Thus, Myr might be a potential strategy for therapy or adjuvant therapy for malignant cardiac hypertrophy.
Collapse
|
54
|
Han X, Wang Y, Fu M, Song Y, Wang J, Cui X, Fan Y, Cao J, Luo J, Sun A, Zou Y, Hu K, Zhou J, Ge J. Effects of Adiponectin on Diastolic Function in Mice Underwent Transverse Aorta Constriction. J Cardiovasc Transl Res 2019; 13:225-237. [PMID: 31621035 PMCID: PMC7166206 DOI: 10.1007/s12265-019-09913-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023]
Abstract
Diastolic dysfunction is common in various cardiovascular diseases, which could be affected by adiponectin (APN). Nevertheless, the effects of APN on diastolic dysfunction in pressure overload model induced by transverse aorta constriction (TAC) remain to be further elucidated. Here, we demonstrated that treatment of APN attenuated diastolic dysfunction and cardiac hypertrophy in TAC mice. Notably, APN also improved active relaxation of adult cardiomyocytes, increased N2BA/N2B ratios of titin isoform, and reduced collagen type I to type III ratio and lysyl oxidase (Lox) expressions in the myocardial tissue. Moreover, APN supplementation suppressed TAC-induced oxidative stress. In vitro, inhibition of AMPK by compound C (Cpc) abrogated the effect of APN on modulation of titin isoform shift and the anti-hypertrophic effect of APN on cardiomyocytes induced by AngII. In summary, our findings indicate that APN could attenuate diastolic dysfunction in TAC mice, which are at least partially mediated by AMPK pathway.
Collapse
Affiliation(s)
- Xueting Han
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanyan Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mingqiang Fu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Song
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingfeng Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaotong Cui
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuyuan Fan
- North Sichuan Medical College, Nanchong, Sichuan, China
| | - Juan Cao
- North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Luo
- North Sichuan Medical College, Nanchong, Sichuan, China
| | - Aijun Sun
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunzeng Zou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kai Hu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingmin Zhou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
55
|
Asiatic acid inhibits cardiac fibrosis throughNrf2/HO-1 and TGF-β1/Smads signaling pathways in spontaneous hypertension rats. Int Immunopharmacol 2019; 74:105712. [DOI: 10.1016/j.intimp.2019.105712] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 05/26/2019] [Accepted: 06/18/2019] [Indexed: 01/13/2023]
|
56
|
Amaral JH, Rizzi ES, Alves-Lopes R, Pinheiro LC, Tostes RC, Tanus-Santos JE. Antioxidant and antihypertensive responses to oral nitrite involves activation of the Nrf2 pathway. Free Radic Biol Med 2019; 141:261-268. [PMID: 31251976 DOI: 10.1016/j.freeradbiomed.2019.06.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/14/2019] [Accepted: 06/24/2019] [Indexed: 12/27/2022]
Abstract
Impaired redox balance contributes to the cardiovascular alterations of hypertension and activation of nuclear factor erythroid 2-related factor 2 (Nrf2) pathway may counteract these alterations. While nitrite recycles back to NO and exerts antioxidant and antihypertensive effects, the mechanisms involved in these responses are not fully understood. We hypothesized that nitrite treatment of two-kidney, one-clip (2K1C) hypertensive rats activates the Nrf2 pathway, promotes the transcription of antioxidant genes, and improves the vascular redox imbalance and dysfunction in this model. Two doses of oral nitrite were studied: 15 mg/kg and the sub-antihypertensive dose of 1 mg/kg. Nitrite 15 mg/kg (but not 1 mg/kg) decreased blood pressure and increased circulating plasma nitrite and nitrate. Both doses blunted hypertension-induced increases in mesenteric artery reactive oxygen species concentrations assessed by DHE technique and restored the impaired mesenteric artery responses to acetylcholine. While 2K1C hypertension decreased nuclear Nrf2 accumulation, both doses of nitrite increased nuclear Nrf2 accumulation and mRNA expression of Nrf2-regulated genes including superoxide dismutase-1 (SOD1), catalase (CAT), glutathione peroxidase (GPX), thioredoxin-1(TRDX-1) and -2 (TRDX-2). To further confirm nitrite-mediated antioxidant effects, we measured vascular SOD and GPX activity and we found that nitrite at 1 or 15 mg/kg increased the activity of both enzymes (P < 0.05). These results suggest that activation of the Nrf2 pathway promotes antioxidant effects of nitrite, which may improve the vascular dysfunction in hypertension, even when nitrite is given at a sub-antihypertensive dose. These findings may have many clinical implications, particularly in the therapy of hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Jefferson H Amaral
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Elen S Rizzi
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Rhéure Alves-Lopes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Lucas C Pinheiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil
| | - Jose E Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirao Preto, SP, Brazil.
| |
Collapse
|
57
|
Zhou S, Jin J, Wang J, Zhang Z, Huang S, Zheng Y, Cai L. Effects of Breast Cancer Genes 1 and 2 on Cardiovascular Diseases. Curr Probl Cardiol 2019; 46:100421. [PMID: 31558344 DOI: 10.1016/j.cpcardiol.2019.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/06/2019] [Indexed: 12/20/2022]
Abstract
Carriers of mutations of breast cancer gene 1 and/or 2 (BRCA1/2) have a higher risk of developing breast and ovarian cancers at a relatively young age. Recently, a causative role for BRCA1/2 in cardiovascular diseases has been emerging. In this review, we summarize currently available evidence obtained from studies on animal models and human BRCA1/2 mutation carriers that shows a correlation of BRCA1/2 deficiency with various cardiovascular diseases, including ischemic heart disease, atherosclerosis, and chemotherapy-linked cardiac muscle disorders. We also discuss one of the major mechanisms by which BRCA1/2 protects the heart against oxidative stress, ie mediating the activity of Nrf2 and its downstream targets that govern antioxidant signaling. More research is needed to elucidate whether the carriers of the BRCA1/2 mutations with ovarian and breast cancers have increased susceptibility to chemotherapy-induced cardiac functional impairment.
Collapse
|
58
|
Ge ZD, Lian Q, Mao X, Xia Z. Current Status and Challenges of NRF2 as a Potential Therapeutic Target for Diabetic Cardiomyopathy. Int Heart J 2019; 60:512-520. [PMID: 30971629 DOI: 10.1536/ihj.18-476] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Diabetic cardiomyopathy is one of the main causes of heart failure and death in patients with diabetes mellitus. Reactive oxygen species produced excessively in diabetes mellitus cause necrosis, apoptosis, ferroptosis, inflammation, and fibrosis of the myocardium as well as impair the cardiac structure and function. It is increasingly clear that oxidative stress is a principal cause of diabetic cardiomyopathy. The transcription factor nuclear factor-erythroid 2 p45-related factor 2 (NRF2) activates the transcription of more than 200 genes in the human genome. Most of the proteins translated from these genes possess anti-oxidant, anti-inflammatory, anti-apoptotic, anti-ferroptotic, and anti-fibrotic actions. There is a growing body of evidence indicating that NRF2 and its target genes are crucial in preventing high glucose-induced oxidative damage in diabetic cardiomyopathy. Recently, many natural and synthetic activators of NRF2 are shown to possess promising therapeutic effects on diabetic cardiomyopathy in animal models of diabetic cardiomyopathy. Targeting NRF2 signaling by pharmacological entities is a potential approach to ameliorating diabetic cardiomyopathy. However, the persistent high expression of NRF2 in cancer tissues also protects the growth of cancer cells. This "dark side" of NRF2 increases the challenges of using NRF2 activators to treat diabetic cardiomyopathy. In addition, some NRF2 activators were found to have off-target effects. In this review, we summarize the current status and challenges of NRF2 as a potential therapeutic target for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Zhi-Dong Ge
- Department of Anesthesiology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou.,Department of Anesthesiology, Medical College of Wisconsin, Milwaukee
| | - Qingquan Lian
- Department of Anesthesiology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou
| | - Xiaowen Mao
- Department of Anesthesiology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou
| | - Zhengyuan Xia
- Department of Anesthesiology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou.,Department of Anesthesiology, The University of Hong Kong
| |
Collapse
|
59
|
Sathibabu Uddandrao VV, Brahmanaidu P, Nivedha PR, Vadivukkarasi S, Saravanan G. Beneficial Role of Some Natural Products to Attenuate the Diabetic Cardiomyopathy Through Nrf2 Pathway in Cell Culture and Animal Models. Cardiovasc Toxicol 2019; 18:199-205. [PMID: 29080123 DOI: 10.1007/s12012-017-9430-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Diabetic cardiomyopathy, as one of the main cardiac complications in diabetic patients, is identified to connect with oxidative stress that is due to interruption in balance between reactive oxygen species or/and reactive nitrogen species generation and their clearance by antioxidant protection systems. Transcription factor the nuclear factor erythroid 2-related factor 2 (Nrf2) plays a significant role in maintaining the oxidative homeostasis by regulating multiple downstream antioxidants. The Nrf2 plays a significant role in ARE-mediated basal and inducible expression of more than 200 genes that can be grouped into numerous categories as well as antioxidant genes and phase II detoxifying enzymes. On the other hand, activation of Nrf2 by natural and synthetic therapeutics or antioxidants has been revealed effective for the prevention and treatment of toxicities and diseases connected with oxidative stress. Hence, recently focus has been shifted toward plants and plant-based medicines in curing such chronic diseases, as they are supposed to be less toxic. In this review, we focused on the role of some natural products on diabetic cardiomyopathy through Nrf2 pathway.
Collapse
Affiliation(s)
- V V Sathibabu Uddandrao
- Department of Biochemistry, Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous), Thokkavadi, Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - Parim Brahmanaidu
- Department of Biotechnology, Vikrama Simhapuri University, Nellore, Andhra Pradesh, 524003, India
| | - P R Nivedha
- Department of Biochemistry, Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous), Thokkavadi, Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - S Vadivukkarasi
- Department of Biochemistry, Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous), Thokkavadi, Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - Ganapathy Saravanan
- Department of Biochemistry, Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous), Thokkavadi, Tiruchengode, Namakkal District, Tamilnadu, 637215, India.
| |
Collapse
|
60
|
Chen D, Li Z, Bao P, Chen M, Zhang M, Yan F, Xu Y, Ji C, Hu X, Sanchis D, Zhang Y, Ye J. Nrf2 deficiency aggravates Angiotensin II-induced cardiac injury by increasing hypertrophy and enhancing IL-6/STAT3-dependent inflammation. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1253-1264. [PMID: 30668979 DOI: 10.1016/j.bbadis.2019.01.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/07/2019] [Accepted: 01/16/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND NF-E2-related factor 2 (Nrf2) is a transcription factor playing cytoprotective effects in various pathological processes including oxidative stress and cardiac hypertrophy. Despite being a potential therapeutic target to treat several cardiomyopathies, the signaling underlying Nrf2-dependent cardioprotective action remains largely uncharacterized. AIM This study aimed to explore the signaling mediating the role of Nrf2 in the development of hypertensive cardiac pathogenesis by analyzing the response to Angiotensin II (Ang II) in the presence or absence of Nrf2 expression, both in vivo and in vitro. RESULTS Our results indicated that Nrf2 deficiency exacerbated cardiac damage triggered by Ang II infusion. Mechanistically, our study shows that Ang II-triggered hypertrophy and inflammation is exacerbated in the absence of Nrf2 expression and points to the involvement of the IL-6/STAT3 signaling pathway in this event. Indeed, our results show that IL-6 abundance triggered by Ang II is increased in the absence of Nrf2 and demonstrate the requirement of IL-6 in STAT3 activation and cardiac inflammation induced by Ang II. CONCLUSION Our results show that Nrf2 is important for the protection of the heart against Ang II-induced cardiac hypertrophy and inflammation by mechanisms involving the regulation of IL-6/STAT3-dependent signaling.
Collapse
Affiliation(s)
- Dandan Chen
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Zhe Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular research Institute, Wuhan University, Wuhan 430060, China; Hubei key Laboratory of Cardiology, Wuhan 430060, China
| | - Peiqing Bao
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Miao Chen
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Miao Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Yitao Xu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London W120NN, United Kingdom
| | - Caoyu Ji
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210006, China
| | - Xinyue Hu
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China
| | - Daniel Sanchis
- Institut de Recerca Biomedica de Lleida (IRBLLEIDA), Universitat de Lleida, Edifici Biomedicina-I. Av. Rovira Roure, 80, 25198 Lleida, Spain.
| | - Yubin Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| | - Junmei Ye
- State Key Laboratory of Natural Medicines, Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210006, China.
| |
Collapse
|
61
|
In-vivo correlations between skin metabolic oscillations and vasomotion in wild-type mice and in a model of oxidative stress. Sci Rep 2019; 9:186. [PMID: 30655574 PMCID: PMC6336806 DOI: 10.1038/s41598-018-36970-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/27/2018] [Indexed: 12/17/2022] Open
Abstract
Arterioles in the cutaneous microcirculation frequently display an oscillatory phenomenon defined vasomotion, consistent with periodic diameter variations in the micro-vessels associated with particular physiological or abnormal conditions. The cellular mechanisms underlying vasomotion and its physiological role have not been completely elucidated. Various mechanisms were demonstrated, based on cell Ca2+ oscillations determined by the activity of channels in the plasma membrane or sarcoplasmic reticulum of vascular cells. However, the possible engagement in vasomotion of cell metabolic oscillations of mitochondrial or glycolytic origin has been poorly explored. Metabolic oscillations associated with the production of ATP energy were previously described in cells, while limited studies have investigated these fluctuations in-vivo. Here, we characterised a low-frequency metabolic oscillator (MO-1) in skin from live wild-type and Nrf2−/− mice, by combination of fluorescence spectroscopy and wavelet transform processing technique. Furthermore, the relationships between metabolic and microvascular oscillators were examined during phenylephrine-induced vasoconstriction. We found a significant interaction between MO-1 and the endothelial EDHF vasomotor mechanism that was reduced in the presence of oxidative stress (Nrf2−/− mice). Our findings suggest indirectly that metabolic oscillations may be involved in the mechanisms underlying endothelium-mediated skin vasomotion, which might be altered in the presence of metabolic disturbance.
Collapse
|
62
|
Spencer Noakes TL, Przybycien TS, Forwell A, Nicholls C, Zhou YQ, Butcher DT, Weksberg R, Guger SL, Spiegler BJ, Schachar RJ, Hitzler J, Ito S, van der Plas E, Nieman BJ. Brain Development and Heart Function after Systemic Single-Agent Chemotherapy in a Mouse Model of Childhood Leukemia Treatment. Clin Cancer Res 2018; 24:6040-6052. [PMID: 30054283 DOI: 10.1158/1078-0432.ccr-18-0551] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/19/2018] [Accepted: 07/24/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Chemotherapy for childhood acute lymphoblastic leukemia (ALL) can cause late-appearing side effects in survivors that affect multiple organs, including the heart and brain. However, the complex ALL treatment regimen makes it difficult to isolate the causes of these side effects and impossible to separate the contributions of individual chemotherapy agents by clinical observation. Using a mouse model, we therefore assessed each of eight representative, systemically-administered ALL chemotherapy agents for their impact on postnatal brain development and heart function. EXPERIMENTAL DESIGN Mice were treated systemically with a single chemotherapy agent at an infant equivalent age, then allowed to age to early adulthood (9 weeks). Cardiac structure and function were assessed using in vivo high-frequency ultrasound, and brain anatomy was assessed using high-resolution volumetric ex vivo MRI. In addition, longitudinal in vivo MRI was used to determine the time course of developmental change after vincristine treatment. RESULTS Vincristine, doxorubicin, and methotrexate were observed to produce the greatest deficiencies in brain development as determined by volumes measured on MRI, whereas doxorubicin, methotrexate, and l-asparaginase altered heart structure or function. Longitudinal studies of vincristine revealed widespread volume loss immediately following treatment and impaired growth over time in several brain regions. CONCLUSIONS Multiple ALL chemotherapy agents can affect postnatal brain development or heart function. This study provides a ranking of agents based on potential toxicity, and thus highlights a subset likely to cause side effects in early adulthood for further study.
Collapse
Affiliation(s)
- T Leigh Spencer Noakes
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Translational Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Thomas S Przybycien
- Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amanda Forwell
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- The University of Waterloo, Waterloo, Ontario, Canada
| | - Connor Nicholls
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- The University of Waterloo, Waterloo, Ontario, Canada
| | - Yu-Qing Zhou
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, The University of Toronto, Ontario, Canada
| | - Darci T Butcher
- Genetics & Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Rosanna Weksberg
- Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Genetics & Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Clinical and Metabolic Genetics, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Molecular Genetics, The University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, The University of Toronto, Toronto, Ontario, Canada
| | - Sharon L Guger
- Department of Molecular Genetics, The University of Toronto, Toronto, Ontario, Canada
| | - Brenda J Spiegler
- Department of Psychology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, Faculty of Medicine, The University of Toronto, Toronto, Ontario, Canada
| | - Russell J Schachar
- Department of Psychiatry, The Hospital for Sick Children, Toronto, Ontario, Canada
- Psychiatry Research, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Johann Hitzler
- Translational Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Pediatrics, Faculty of Medicine, The University of Toronto, Toronto, Ontario, Canada
- Development and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Shinya Ito
- Translational Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Clinical Pharmacology and Toxicology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Pharmacology and Toxicology, Faculty of Medicine, The University of Toronto, Toronto, Ontario, Canada
| | - Ellen van der Plas
- Department of Psychiatry, The University of Iowa Hospital and Clinics, Iowa City, Iowa
| | - Brian J Nieman
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
63
|
Kazakov A, Hall RA, Werner C, Meier T, Trouvain A, Rodionycheva S, Nickel A, Lammert F, Maack C, Böhm M, Laufs U. Raf kinase inhibitor protein mediates myocardial fibrosis under conditions of enhanced myocardial oxidative stress. Basic Res Cardiol 2018; 113:42. [PMID: 30191336 PMCID: PMC6133069 DOI: 10.1007/s00395-018-0700-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/15/2018] [Indexed: 12/21/2022]
Abstract
Fibrosis is a hallmark of maladaptive cardiac remodelling. Here we report that genome-wide quantitative trait locus (QTL) analyses in recombinant inbred mouse lines of C57BL/6 J and DBA2/J strains identified Raf Kinase Inhibitor Protein (RKIP) as genetic marker of fibrosis progression. C57BL/6 N-RKIP−/− mice demonstrated diminished fibrosis induced by transverse aortic constriction (TAC) or CCl4 (carbon tetrachloride) treatment compared with wild-type controls. TAC-induced expression of collagen Iα2 mRNA, Ki67+ fibroblasts and marker of oxidative stress 8-hydroxyguanosine (8-dOHG)+ fibroblasts as well as the number of fibrocytes in the peripheral blood and bone marrow were markedly reduced in C57BL/6 N-RKIP−/− mice. RKIP-deficient cardiac fibroblasts demonstrated decreased migration and fibronectin production. This was accompanied by a two-fold increase of the nuclear accumulation of nuclear factor erythroid 2-related factor 2 (Nrf2), the main transcriptional activator of antioxidative proteins, and reduced expression of its inactivators. To test the importance of oxidative stress for this signaling, C57BL/6 J mice were studied. C57BL/6 J, but not the C57BL/6 N-strain, is protected from TAC-induced oxidative stress due to mutation of the nicotinamide nucleotide transhydrogenase gene (Nnt). After TAC surgery, the hearts of Nnt-deficient C57BL/6 J-RKIP−/− mice revealed diminished oxidative stress, increased left ventricular (LV) fibrosis and collagen Iα2 as well as enhanced basal nuclear expression of Nrf2. In human LV myocardium from both non-failing and failing hearts, RKIP-protein correlated negatively with the nuclear accumulation of Nrf2. In summary, under conditions of Nnt-dependent enhanced myocardial oxidative stress induced by TAC, RKIP plays a maladaptive role for fibrotic myocardial remodeling by suppressing the Nrf2-related beneficial effects.
Collapse
Affiliation(s)
- Andrey Kazakov
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany.
| | - Rabea A Hall
- Klinik für Innere Medizin II, Gastroenterologie, Hepatologie, Endokrinologie, Diabetologie und Ernährungsmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 77, 66421, Homburg, Germany
| | - Christian Werner
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany
| | - Timo Meier
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany
| | - André Trouvain
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany
| | - Svetlana Rodionycheva
- Klinik für Thorax- und Herz-Gefäßchirurgie, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 57, 66421, Homburg, Germany
| | - Alexander Nickel
- Deutsches Zentrum für Herzinsuffizienz, Universitätsklinikum Würzburg, am Schwarzenberg 15, A15, 97078, Würzburg, Germany
| | - Frank Lammert
- Klinik für Innere Medizin II, Gastroenterologie, Hepatologie, Endokrinologie, Diabetologie und Ernährungsmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 77, 66421, Homburg, Germany
| | - Christoph Maack
- Deutsches Zentrum für Herzinsuffizienz, Universitätsklinikum Würzburg, am Schwarzenberg 15, A15, 97078, Würzburg, Germany
| | - Michael Böhm
- Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Universität/Universitätsklinikum des Saarlandes, Kirrberger Strasse 100, IMED, 66421, Homburg, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Liebigstrasse 20, 04103, Leipzig, Germany
| |
Collapse
|
64
|
Zhao GJ, Hou N, Cai SA, Liu XW, Li AQ, Cheng CF, Huang Y, Li LR, Mai YP, Liu SM, Ou CW, Xiong ZY, Chen XH, Chen MS, Luo CF. Contributions of Nrf2 to Puerarin Prevention of Cardiac Hypertrophy and its Metabolic Enzymes Expression in Rats. J Pharmacol Exp Ther 2018; 366:458-469. [PMID: 29945930 DOI: 10.1124/jpet.118.248369] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/21/2018] [Indexed: 12/16/2022] Open
Abstract
Previous evidence has suggested that puerarin may attenuate cardiac hypertrophy; however, the potential mechanisms have not been determined. Moreover, the use of puerarin is limited by severe adverse events, including intravascular hemolysis. This study used a rat model of abdominal aortic constriction (AAC)-induced cardiac hypertrophy to evaluate the potential mechanisms underlying the attenuating efficacy of puerarin on cardiac hypertrophy, as well as the metabolic mechanisms of puerarin involved. We confirmed that puerarin (50 mg/kg per day) significantly attenuated cardiac hypertrophy, upregulated Nrf2, and decreased Keap1 in the myocardium. Moreover, puerarin significantly promoted Nrf2 nuclear accumulation in parallel with the upregulated downstream proteins, including heme oxygenase 1, glutathione transferase P1, and NAD(P)H:quinone oxidoreductase 1. Similar results were obtained in neonatal rat cardiomyocytes (NRCMs) treated with angiotensin II (Ang II; 1 μM) and puerarin (100 μM), whereas the silencing of Nrf2 abolished the antihypertrophic effects of puerarin. The mRNA and protein levels of UGT1A1 and UGT1A9, enzymes for puerarin metabolism, were significantly increased in the liver and heart tissues of AAC rats and Ang II-treated NRCMs. Interestingly, the silencing of Nrf2 attenuated the puerarin-induced upregulation of UGT1A1 and UGT1A9. The results of chromatin immunoprecipitation-quantitative polymerase chain reaction indicated that the binding of Nrf2 to the promoter region of Ugt1a1 or Ugt1a9 was significantly enhanced in puerarin-treated cardiomyocytes. These results suggest that Nrf2 is the key regulator of antihypertrophic effects and upregulation of the metabolic enzymes UGT1A1 and UGT1A9 of puerarin. The autoregulatory circuits between puerarin and Nrf2-induced UGT1A1/1A9 are beneficial to attenuate adverse effects and maintain the pharmacologic effects of puerarin.
Collapse
Affiliation(s)
- Gan-Jian Zhao
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Ning Hou
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Shao-Ai Cai
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Xia-Wen Liu
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Ai-Qun Li
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Chuan-Fang Cheng
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Yin Huang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Li-Rong Li
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Yun-Pei Mai
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Shi-Ming Liu
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Cai-Wen Ou
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Zhen-Yu Xiong
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Xiao-Hui Chen
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Min-Sheng Chen
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| | - Cheng-Feng Luo
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Cardiovascular Disease (G.-J.Z., A.-Q.L., C.-F.C., Y.H., L.-R.L., S.-M.L., C.-F.L.), School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University (N.H., X.-W.L., Y.-P.M.), The Second Affiliated Hospital of Guangzhou Medical University (S.-A.C., X.-H.C.); Zhujiang Hospital, Southern Medical University, Guangdong Provincial Center of Biomedical Engineering for Cardiovascular Disease (C.-W.O., M.-S.C.), and The First Affiliated Hospital, Sun Yat-sen University (Z.-Y.X.), Guangzhou, China
| |
Collapse
|
65
|
Diederich L, Suvorava T, Sansone R, Keller TCS, Barbarino F, Sutton TR, Kramer CM, Lückstädt W, Isakson BE, Gohlke H, Feelisch M, Kelm M, Cortese-Krott MM. On the Effects of Reactive Oxygen Species and Nitric Oxide on Red Blood Cell Deformability. Front Physiol 2018; 9:332. [PMID: 29867516 PMCID: PMC5958211 DOI: 10.3389/fphys.2018.00332] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/16/2018] [Indexed: 01/08/2023] Open
Abstract
The main function of red blood cells (RBCs) is the transport of respiratory gases along the vascular tree. To fulfill their task, RBCs are able to elastically deform in response to mechanical forces and, pass through the narrow vessels of the microcirculation. Decreased RBC deformability was observed in pathological conditions linked to increased oxidative stress or decreased nitric oxide (NO) bioavailability, like hypertension. Treatments with oxidants and with NO were shown to affect RBC deformability ex vivo, but the mechanisms underpinning these effects are unknown. In this study we investigate whether changes in intracellular redox status/oxidative stress or nitrosation reactions induced by reactive oxygen species (ROS) or NO may affect RBC deformability. In a case-control study comparing RBCs from healthy and hypertensive participants, we found that RBC deformability was decreased, and levels of ROS were increased in RBCs from hypertensive patients as compared to RBCs from aged-matched healthy controls, while NO levels in RBCs were not significantly different. To study the effects of oxidants on RBC redox state and deformability, RBCs from healthy volunteers were treated with increasing concentrations of tert-butylhydroperoxide (t-BuOOH). We found that high concentrations of t-BuOOH (≥ 1 mM) significantly decreased the GSH/GSSG ratio in RBCs, decreased RBC deformability and increased blood bulk viscosity. Moreover, RBCs from Nrf2 knockout (KO) mice, a strain genetically deficient in a number of antioxidant/reducing enzymes, were more susceptible to t-BuOOH-induced impairment in RBC deformability as compared to wild type (WT) mice. To study the role of NO in RBC deformability we treated RBC suspensions from human volunteers with NO donors and nitrosothiols and analyzed deformability of RBCs from mice lacking the endothelial NO synthase (eNOS). We found that NO donors induced S-nitrosation of the cytoskeletal protein spectrin, but did not affect human RBC deformability or blood bulk viscosity; moreover, under unstressed conditions RBCs from eNOS KO mice showed fully preserved RBC deformability as compared to WT mice. Pre-treatment of human RBCs with nitrosothiols rescued t-BuOOH-mediated loss of RBC deformability. Taken together, these findings suggest that NO does not affect RBC deformability per se, but preserves RBC deformability in conditions of oxidative stress.
Collapse
Affiliation(s)
- Lukas Diederich
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Tatsiana Suvorava
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Roberto Sansone
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - T C Stevenson Keller
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Frederik Barbarino
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Thomas R Sutton
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Christian M Kramer
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Wiebke Lückstädt
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Brant E Isakson
- Department of Molecular Physiology and Biological Physics, Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Holger Gohlke
- Faculty of Mathematics and Natural Sciences, Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Malte Kelm
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Medical Faculty, Cardiovascular Research Institute Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
66
|
Hydrogen Gas Protects Against Intestinal Injury in Wild Type But Not NRF2 Knockout Mice With Severe Sepsis by Regulating HO-1 and HMGB1 Release. Shock 2018; 48:364-370. [PMID: 28234792 DOI: 10.1097/shk.0000000000000856] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The intestine plays an important role in the pathogenesis of sepsis. Hydrogen gas (H2), which has anti-oxidative, anti-inflammatory, and anti-apoptotic effects, can be effectively used to treat septic mice. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a redox-sensitive master switch that regulates the expression of antioxidant and protective enzymes. This study investigated the effects of 2% H2 on intestinal injuries and the underlying mechanisms in a mouse model of severe sepsis. Male Nrf2 knockout mice (Nrf2-KO) and wild-type (WT) mice were randomized into four groups: sham, sham+H2, cecal ligation and puncture (CLP), and CLP+H2. The survival rate was observed and recorded within 7 days, and pro-inflammatory cytokines (TNF-α, IL-6, HMGB1), anti-inflammatory cytokine (IL-10), antioxidant enzymes (superoxide dismutase, and catalase ), and oxidative products (MDA, 8-iso-PGF2α) were detected in the serum and intestine using an enzyme-linked immunosorbent assay. In addition, the protein and mRNA levels of heme oxygenase-1 (HO-1) and high mobility group box 1 (HMGB1) were measured by Western blotting and quantitative PCR, respectively. Immunofluorescence and immunohistochemistry were used to measure HMGB1 and HO-1 release into the intestine, respectively. The results showed that therapy with 2% H2 increased the survival rate, alleviated the injuries caused by oxidative stress and inflammation, reduced HMGB1 levels but increased HO-1 levels in WT septic mice, but not in Nrf2-KO mice. These data demonstrate that 2% H2 inhalation may be a promising therapeutic strategy for intestinal injuries caused by severe sepsis through the regulation of HO-1 and HMGB1 release. In addition, Nrf2 plays a key role in the protective effects of H2 against intestinal damage in this disease.
Collapse
|
67
|
Nrf2 Deficiency Unmasks the Significance of Nitric Oxide Synthase Activity for Cardioprotection. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8309698. [PMID: 29854098 PMCID: PMC5952436 DOI: 10.1155/2018/8309698] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/17/2018] [Accepted: 02/27/2018] [Indexed: 12/14/2022]
Abstract
The transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a key master switch that controls the expression of antioxidant and cytoprotective enzymes, including enzymes catalyzing glutathione de novo synthesis. In this study, we aimed to analyze whether Nrf2 deficiency influences antioxidative capacity, redox state, NO metabolites, and outcome of myocardial ischemia reperfusion (I/R) injury. In Nrf2 knockout (Nrf2 KO) mice, we found elevated eNOS expression and preserved NO metabolite concentrations in the aorta and heart as compared to wild types (WT). Unexpectedly, Nrf2 KO mice have a smaller infarct size following myocardial ischemia/reperfusion injury than WT mice and show fully preserved left ventricular systolic function. Inhibition of NO synthesis at onset of ischemia and during early reperfusion increased myocardial damage and systolic dysfunction in Nrf2 KO mice, but not in WT mice. Consistent with this, infarct size and diastolic function were unaffected in eNOS knockout (eNOS KO) mice after ischemia/reperfusion. Taken together, these data suggest that eNOS upregulation under conditions of decreased antioxidant capacity might play an important role in cardioprotection against I/R. Due to the redundancy in cytoprotective mechanisms, this fundamental antioxidant property of eNOS is not evident upon acute NOS inhibition in WT mice or in eNOS KO mice until Nrf2-related signaling is abrogated.
Collapse
|
68
|
Wang G, Li Y, Peng Y, Tang J, Li H. Association of polymorphisms in MALAT1 with risk of coronary atherosclerotic heart disease in a Chinese population. Lipids Health Dis 2018; 17:75. [PMID: 29631611 PMCID: PMC5891990 DOI: 10.1186/s12944-018-0728-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 03/28/2018] [Indexed: 12/13/2022] Open
Abstract
Background Metastasis associated lung adenocarcinoma transcript 1 (MALAT1) plays an important role in vascular remodeling. Down-regulation of MALAT1 can inhibit the proliferation of vascular endothelial cells and vascular smooth muscle cells, reduce cardiomyocyte apoptosis and improve left ventricular function, which is closely linked to numerous pathological processes such as coronary atherosclerotic heart disease (CAD). The aim of this study was to investigate whether polymorphisms in MALAT1 were associated with the susceptibility to CAD. Methods A total of 508 CAD patients and 562 age-, gender-, and ethnicity-matched controls were enrolled in this study. Four polymorphisms in MALAT1 (i.e., rs11227209, rs619586, rs664589, and rs3200401) were genotyped using a TaqMan allelic discrimination assay. Results The rs619586 AG/GG genotypes and G allele were associated with a reduced risk of CAD (AG/GG vs. AA: adjusted OR = 0.66, 95% CI: 0.48–0.91; G vs. A: adjusted OR = 0.68, 95% CI: 0.51–0.90). Stratification analyses showed that CAD patients with rs11227209 CG/GG, rs619586 AG/GG, and rs3200401 CT/TT genotypes exhibited lower levels of TCH (P = 0.02, 0.04, and 0.02, respectively). Moreover, CGCC haplotype was associated with a decreased risk of CAD (OR = 0.28, 95% CI: 0.16–0.48). Multivariate logistic regression analysis identified some independent risk factors for CAD, including rs619586 and rs664589. Subsequent combined analysis showed that the combined genotypes of rs619586AG/GG and rs664589CC were associated with a reduced risk of CAD (OR = 0.29; 95%CI, 0.16–0.53). Conclusions These findings indicate that rs619586AG/GG genotypes in MALAT1 may protect against the occurrence of CAD. Electronic supplementary material The online version of this article (10.1186/s12944-018-0728-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Genan Wang
- Department of Heart Vascular Surgery, Yan'An Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, People's Republic of China
| | - Yaxiong Li
- Department of Heart Vascular Surgery, Yan'An Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, People's Republic of China
| | - Yong Peng
- Department of Heart Vascular Surgery, Yan'An Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, People's Republic of China
| | - Jian Tang
- Department of Heart Vascular Surgery, Yan'An Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, People's Republic of China
| | - Hua Li
- Department of Heart Vascular Surgery, Yan'An Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, People's Republic of China.
| |
Collapse
|
69
|
Abstract
There are multiple intrinsic mechanisms for diastolic dysfunction ranging from molecular to structural derangements in ventricular myocardium. The molecular mechanisms regulating the progression from normal diastolic function to severe dysfunction still remain poorly understood. Recent studies suggest a potentially important role of core cardio-enriched transcription factors (TFs) in the control of cardiac diastolic function in health and disease through their ability to regulate the expression of target genes involved in the process of adaptive and maladaptive cardiac remodeling. The current relevant findings on the role of a variety of such TFs (TBX5, GATA-4/6, SRF, MYOCD, NRF2, and PITX2) in cardiac diastolic dysfunction and failure are updated, emphasizing their potential as promising targets for novel treatment strategies. In turn, the new animal models described here will be key tools in determining the underlying molecular mechanisms of disease. Since diastolic dysfunction is regulated by various TFs, which are also involved in cross talk with each other, there is a need for more in-depth research from a biomedical perspective in order to establish efficient therapeutic strategies.
Collapse
|
70
|
Rosuvastatin improves myocardial hypertrophy after hemodynamic pressure overload via regulating the crosstalk of Nrf2/ARE and TGF-β/ smads pathways in rat heart. Eur J Pharmacol 2017; 820:173-182. [PMID: 29225188 DOI: 10.1016/j.ejphar.2017.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 11/07/2017] [Accepted: 12/06/2017] [Indexed: 01/09/2023]
Abstract
Left ventricular hypertrophy is more commonly associated with hemodynamic overload imposed by hypertension or volume overload. Transforming growth factor β (TGF-β) is involved in the cardiac hypertrophy and fibrosis of the left ventricle. The fact that TGF-β1 and the nuclear factor erythroid 2-related factor 2 (Nrf2) both become up-regulated upon persistent vessel overload suggests that these two factors may virtually impact on their signaling pathways. In this research, 40 rats were divided into sham group, model group, rosuvastatin low and high dose group. Rat models were established by incomplete constriction of abdominal aorta. After five weeks treatment, blood pressure, heart mass index (HMI), hemodynamic parameters and the average diameter of myocardium cell and collagen volume fraction (CVF) improved significantly in rosuvastatin groups, compared with the model group. Both rosuvastatin groups, increased in expression of Smad7, Nrf2, NAD (P) H dehydrogenase [quinone] 1 (Nqo1) and heme oxygenase 1(Ho1),and decreased in expression of TGF-βl、Smad3 compared with the model group. Results from co-immunoprecipitation and GST pull down showed that Nrf2 interacts with Smad7. Our results revealed the crosstalk between TGF-β1/Smads and Nrf2/ antioxidant response elements (ARE) pathways in myocardial remodeling through the interaction between Smad7 and Nrf2. Rosuvastatin can improve cardiac function and hypertrophy by regulating the crosstalk of the two signaling pathways.
Collapse
|
71
|
Schnelle M, Catibog N, Zhang M, Nabeebaccus AA, Anderson G, Richards DA, Sawyer G, Zhang X, Toischer K, Hasenfuss G, Monaghan MJ, Shah AM. Echocardiographic evaluation of diastolic function in mouse models of heart disease. J Mol Cell Cardiol 2017; 114:20-28. [PMID: 29055654 PMCID: PMC5807035 DOI: 10.1016/j.yjmcc.2017.10.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/05/2017] [Accepted: 10/18/2017] [Indexed: 01/08/2023]
Abstract
Background Mouse models of heart disease are extensively employed. The echocardiographic characterization of contractile function is usually focused on systolic function with fewer studies assessing diastolic function. Furthermore, the applicability of diverse echocardiographic parameters of diastolic function that are commonly used in humans has not been extensively evaluated in different pathophysiological models in mice. Methods and results We used high resolution echocardiography to evaluate parameters of diastolic function in mouse models of chronic pressure overload (aortic constriction), volume overload (aorto-caval shunt), heart failure with preserved ejection fraction (HFpEF; DOCA-salt hypertension), and acute sarcoplasmic reticulum dysfunction induced by thapsigargin - all known to exhibit diastolic dysfunction. Left atrial area increased in all three chronic models while mitral E/A was difficult to quantify at high heart rates. Isovolumic relaxation time (IVRT) and Doppler E/E′ increased significantly and the peak longitudinal strain rate during early filling (peak reverse longitudinal strain rate) decreased significantly after aortic constriction, with the changes being proportional to the magnitude of hypertrophy. In the HFpEF model, reverse longitudinal strain rate decreased significantly but changes in IVRT and E/E′ were non-significant, consistent with less severe dysfunction. With volume overload, there was a significant increase in reverse longitudinal strain rate and decrease in IVRT, indicating a restrictive physiology. Acute thapsigargin treatment caused significant prolongation of IVRT and decrease in reverse longitudinal strain rate. Conclusion These results indicate that the combined measurement of left atrial area plus reverse longitudinal strain rate and/or IVRT provide an excellent overall assessment of diastolic function in the diseased mouse heart, allowing distinction between different types of pathophysiology. Several echocardiographic indices of diastolic function are applicable to mouse models. Isovolumic relaxation time (IVRT) and peak strain during filling are easily quantified. Left atrial area increases with pressure and volume overload as well as HFpEF. Changes in IVRT and strain during filling distinguish restrictive physiology. Combined left atrial area and diastolic strain provide an ideal diagnostic framework.
Collapse
Affiliation(s)
- Moritz Schnelle
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom; Department of Cardiology and Pneumology, University Medical Center Goettingen, Goettingen, Germany
| | - Norman Catibog
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom
| | - Min Zhang
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom
| | - Adam A Nabeebaccus
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom
| | - Grace Anderson
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom
| | - Daniel A Richards
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom
| | - Greta Sawyer
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom
| | - Xiaohong Zhang
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom
| | - Karl Toischer
- Department of Cardiology and Pneumology, University Medical Center Goettingen, Goettingen, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University Medical Center Goettingen, Goettingen, Germany
| | - Mark J Monaghan
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, Cardiovascular Division, London, United Kingdom.
| |
Collapse
|
72
|
Wen JJ, Porter C, Garg NJ. Inhibition of NFE2L2-Antioxidant Response Element Pathway by Mitochondrial Reactive Oxygen Species Contributes to Development of Cardiomyopathy and Left Ventricular Dysfunction in Chagas Disease. Antioxid Redox Signal 2017; 27:550-566. [PMID: 28132522 PMCID: PMC5567598 DOI: 10.1089/ars.2016.6831] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIMS We investigated the effects of mitochondrial reactive oxygen species (mtROS) on nuclear factor (erythroid 2)-like 2 (NFE2L2) transcription factor activity during Trypanosoma cruzi (Tc) infection and determined whether enhancing the mtROS scavenging capacity preserved the heart function in Chagas disease. RESULTS C57BL/6 wild type (WT, female) mice infected with Tc exhibited myocardial loss of mitochondrial membrane potential, complex II (CII)-driven coupled respiration, and ninefold increase in mtROS production. In vitro and in vivo studies showed that Tc infection resulted in an ROS-dependent decline in the expression, nuclear translocation, antioxidant response element (ARE) binding, and activity of NFE2L2, and 35-99% decline in antioxidants' (gamma-glutamyl cysteine synthase [γGCS], heme oxygenase-1 [HO1], glutamate-cysteine ligase modifier subunit [GCLM], thioredoxin (Trx), glutathione S transferase [GST], and NAD(P)H dehydrogenase, quinone 1 [NQO1]) expression. An increase in myocardial and mitochondrial oxidative adducts, myocardial interventricular septum thickness, and left ventricle (LV) mass, a decline in LV posterior wall thickness, and disproportionate synthesis of collagens (COLI/COLIII), αSMA, and SM22α were noted in WT.Tc mice. Overexpression of manganese superoxide dismutase (MnSOD) in cultured cells (HeLa or cardiomyocytes) and MnSODtg mice preserved the NFE2L2 transcriptional activity and antioxidant/oxidant balance, and cardiac oxidative and fibrotic pathology were significantly decreased in MnSODtg.Tc mice. Importantly, echocardiography finding of a decline in LV systolic (stroke volume, cardiac output, ejection fraction) and diastolic (early/late peak filling ratio, myocardial performance index) function in WT.Tc mice was abolished in MnSODtg.Tc mice. Innovation and Conclusion: The mtROS inhibition of NFE2L2/ARE pathway constitutes a key mechanism in signaling the fibrotic gene expression and evolution of chronic cardiomyopathy. Preserving the NFE2L2 activity arrested the mitochondrial and cardiac oxidative stress, cardiac fibrosis, and heart failure in Chagas disease. Antioxid. Redox Signal. 27, 550-566.
Collapse
Affiliation(s)
- Jake Jianjun Wen
- 1 Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB) , Galveston, Texas
| | - Craig Porter
- 2 Metabolism Unit, Shriners Hospital for Children , Galveston, Texas.,3 Department of Surgery, University of Texas Medical Branch (UTMB) , Galveston, Texas
| | - Nisha Jain Garg
- 1 Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB) , Galveston, Texas.,4 Department of Pathology, University of Texas Medical Branch (UTMB) , Galveston, Texas.,5 Institute for Human Infections and Immunity, University of Texas Medical Branch (UTMB) , Galveston, Texas
| |
Collapse
|
73
|
Cortese-Krott MM, Mergia E, Kramer CM, Lückstädt W, Yang J, Wolff G, Panknin C, Bracht T, Sitek B, Pernow J, Stasch JP, Feelisch M, Koesling D, Kelm M. Identification of a soluble guanylate cyclase in RBCs: preserved activity in patients with coronary artery disease. Redox Biol 2017; 14:328-337. [PMID: 29024896 PMCID: PMC5975213 DOI: 10.1016/j.redox.2017.08.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/11/2017] [Accepted: 08/17/2017] [Indexed: 12/21/2022] Open
Abstract
Endothelial dysfunction is associated with decreased NO bioavailability and impaired activation of the NO receptor soluble guanylate cyclase (sGC) in the vasculature and in platelets. Red blood cells (RBCs) are known to produce NO under hypoxic and normoxic conditions; however evidence of expression and/or activity of sGC and downstream signaling pathway including phopshodiesterase (PDE)-5 and protein kinase G (PKG) in RBCs is still controversial. In the present study, we aimed to investigate whether RBCs carry a functional sGC signaling pathway and to address whether this pathway is compromised in coronary artery disease (CAD). Using two independent chromatographic procedures, we here demonstrate that human and murine RBCs carry a catalytically active α1β1-sGC (isoform 1), which converts 32P-GTP into 32P-cGMP, as well as PDE5 and PKG. Specific sGC stimulation by NO+BAY 41-2272 increases intracellular cGMP-levels up to 1000-fold with concomitant activation of the canonical PKG/VASP-signaling pathway. This response to NO is blunted in α1-sGC knockout (KO) RBCs, but fully preserved in α2-sGC KO. In patients with stable CAD and endothelial dysfunction red cell eNOS expression is decreased as compared to aged-matched controls; by contrast, red cell sGC expression/activity and responsiveness to NO are fully preserved, although sGC oxidation is increased in both groups. Collectively, our data demonstrate that an intact sGC/PDE5/PKG-dependent signaling pathway exists in RBCs, which remains fully responsive to NO and sGC stimulators/activators in patients with endothelial dysfunction. Targeting this pathway may be helpful in diseases with NO deficiency in the microcirculation like sickle cell anemia, pulmonary hypertension, and heart failure.
Collapse
Affiliation(s)
- Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany.
| | - Evanthia Mergia
- Institute for Pharmacology and Toxicology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Christian M Kramer
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| | - Wiebke Lückstädt
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| | - Jiangning Yang
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Karolinska Universitetssjukhuset, Solna, 171 76 Stockholm, Sweden
| | - Georg Wolff
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| | - Christina Panknin
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| | - Thilo Bracht
- Medizinisches Proteom-Center, Ruhr- University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Barbara Sitek
- Medizinisches Proteom-Center, Ruhr- University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - John Pernow
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Karolinska Universitetssjukhuset, Solna, 171 76 Stockholm, Sweden
| | - Johannes-Peter Stasch
- Bayer Pharma AG, Aprather Weg 18a, 42096 Wuppertal, Germany; Institute of Pharmacy, University Halle-Wittenberg, Wolfgang-Langenbeck-Strasse 4, 06120 Halle (Saale), Germany
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Tremona Road, SO166YD Southampton, United Kingdom
| | - Doris Koesling
- Institute for Pharmacology and Toxicology, Ruhr-University Bochum, Universitätsstraße 150, 44801 Bochum, Germany
| | - Malte Kelm
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225 Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich Heine University, Moorensstraße 5, 40225 Düsseldorf, Germany
| |
Collapse
|
74
|
Erkens R, Suvorava T, Kramer CM, Diederich LD, Kelm M, Cortese-Krott MM. Modulation of Local and Systemic Heterocellular Communication by Mechanical Forces: A Role of Endothelial Nitric Oxide Synthase. Antioxid Redox Signal 2017; 26:917-935. [PMID: 27927026 PMCID: PMC5455615 DOI: 10.1089/ars.2016.6904] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this review, we discuss the role of nitric oxide (NO) as a key physiological mechanotransducer modulating both local and systemic heterocellular communication and contributing to the integrated (patho)physiology of the cardiovascular system. A deeper understanding of mechanotransduction-mediated local and systemic nodes controlling heterocellular communication between the endothelium, blood cells, and other cell types (e.g., cardiomyocytes) may suggest novel therapeutic strategies for endothelial dysfunction and cardiovascular disease. Recent Advances: Mechanical forces acting on mechanoreceptors on endothelial cells activate the endothelial NO synthase (eNOS) to produce NO. NO participates in (i) abluminal heterocellular communication, inducing vasorelaxation, and thereby regulating vascular tone and blood pressure; (ii) luminal heterocellular communication, inhibiting platelet aggregation, and controlling hemostasis; and (iii) systemic heterocellular communication, contributing to adaptive physiological processes in response to exercise and remote ischemic preconditioning. Interestingly, shear-induced eNOS-dependent activation of vascular heterocellular communication constitutes the molecular basis of all methods applied in the clinical routine for evaluation of endothelial function. Critical Issues and Future Directions: The integrated physiology of heterocellular communication is still not fully understood. Dedicated experimental models are needed to analyze messengers and mechanisms underpinning heterocellular communication in response to physical forces in the cardiovascular system (and elsewhere). Antioxid. Redox Signal. 26, 917-935.
Collapse
Affiliation(s)
- Ralf Erkens
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Tatsiana Suvorava
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Christian M Kramer
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Lukas D Diederich
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Malte Kelm
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf , Düsseldorf, Germany
| |
Collapse
|
75
|
Jakobs P, Serbulea V, Leitinger N, Eckers A, Haendeler J. Nuclear Factor (Erythroid-Derived 2)-Like 2 and Thioredoxin-1 in Atherosclerosis and Ischemia/Reperfusion Injury in the Heart. Antioxid Redox Signal 2017; 26:630-644. [PMID: 27923281 PMCID: PMC5397216 DOI: 10.1089/ars.2016.6795] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 11/08/2016] [Accepted: 12/05/2016] [Indexed: 01/04/2023]
Abstract
SIGNIFICANCE Redox signaling is one of the key elements involved in cardiovascular diseases. Two important molecules are the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and the oxidoreductase thioredoxin-1 (Trx-1). Recent Advances: During the previous years, a lot of studies investigated Nrf2 and Trx-1 as protective proteins in cardiovascular disorders. Moreover, post-translational modifications of those molecules were identified that play an important role in the cardiovascular system. This review will summarize changes in the vasculature in atherosclerosis and ischemia reperfusion injury of the heart and the newest findings achieved with Nrf2 and Trx-1 therein. Interestingly, Nrf2 and Trx-1 can act together as well as independently of each other in protection against atherosclerosis and ischemia and reperfusion injury. CRITICAL ISSUES In principle, pharmacological activation of a transcription factor-like Nrf2 can be dangerous, since a transcription regulator has multiple targets and the pleiotropic effects of such activation should not be ignored. Moreover, overactivation of Nrf2 as well as long-term treatment with Trx-1 could be deleterious for the cardiovascular system. FUTURE DIRECTIONS Therefore, the length of treatment with Nrf2 activators and/or Trx-1 has first to be studied in more detail in cardiovascular disorders. Moreover, a combination of Nrf2 activators and Trx-1 should be investigated and taken into consideration. Antioxid. Redox Signal. 26, 630-644.
Collapse
Affiliation(s)
- Philipp Jakobs
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Vlad Serbulea
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
- Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia
| | - Anna Eckers
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Judith Haendeler
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
- Central Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, University of Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
76
|
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
|
77
|
Slezák J, Kura B, Frimmel K, Zálešák M, Ravingerová T, Viczenczová C, Okruhlicová Ľ, Tribulová N. Preventive and therapeutic application of molecular hydrogen in situations with excessive production of free radicals. Physiol Res 2017; 65 Suppl 1:S11-28. [PMID: 27643933 DOI: 10.33549/physiolres.933414] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Excessive production of oxygen free radicals has been regarded as a causative common denominator of many pathological processes in the animal kingdom. Hydroxyl and nitrosyl radicals represent the major cause of the destruction of biomolecules either by a direct reaction or by triggering a chain reaction of free radicals. Scavenging of free radicals may act preventively or therapeutically. A number of substances that preferentially react with free radicals can serve as scavengers, thus increasing the internal capacity/activity of endogenous antioxidants and protecting cells and tissues against oxidative damage. Molecular hydrogen (H(2)) reacts with strong oxidants, such as hydroxyl and nitrosyl radicals, in the cells, that enables utilization of its potential for preventive and therapeutic applications. H(2) rapidly diffuses into tissues and cells without affecting metabolic redox reactions and signaling reactive species. H(2) reduces oxidative stress also by regulating gene expression, and functions as an anti-inflammatory and anti-apoptotic agent. There is a growing body of evidence based on the results of animal experiments and clinical observations that H(2) may represent an effective antioxidant for the prevention of oxidative stress-related diseases. Application of molecular hydrogen in situations with excessive production of free radicals, in particular, hydroxyl and nitrosyl radicals is relatively simple and effective, therefore, it deserves special attention.
Collapse
Affiliation(s)
- J Slezák
- Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovakia.
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Carruth ED, McCulloch AD, Omens JH. Transmural gradients of myocardial structure and mechanics: Implications for fiber stress and strain in pressure overload. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 122:215-226. [PMID: 27845176 DOI: 10.1016/j.pbiomolbio.2016.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although a truly complete understanding of whole heart activation, contraction, and deformation is well beyond our current reach, a significant amount of effort has been devoted to discovering and understanding the mechanisms by which myocardial structure determines cardiac function to better treat patients with cardiac disease. Several experimental studies have shown that transmural fiber strain is relatively uniform in both diastole and systole, in contrast to predictions from traditional mechanical theory. Similarly, mathematical models have largely predicted uniform fiber stress across the wall. The development of this uniform pattern of fiber stress and strain during filling and ejection is due to heterogeneous transmural distributions of several myocardial structures. This review summarizes these transmural gradients, their contributions to fiber mechanics, and the potential functional effects of their remodeling during pressure overload hypertrophy.
Collapse
Affiliation(s)
- Eric D Carruth
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA; Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
79
|
Machin DR, Leary ME, He Y, Shiu YT, Tanaka H, Donato AJ. Ultrasound Assessment of Flow-Mediated Dilation of the Brachial and Superficial Femoral Arteries in Rats. J Vis Exp 2016. [PMID: 27842366 DOI: 10.3791/54762] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Arterial vasodilation to increases in wall shear rate is indicative of vascular endothelial function. In humans, the non-invasive measurement of endothelial function can be achieved by employing the flow-mediated dilation technique, typically performed in the brachial or superficial femoral artery. Briefly, a blood pressure cuff placed distal to an ultrasound probe is inflated to a suprasystolic pressure, which results in limb ischemia. After 5 min of occlusion the cuff is deflated, resulting in reactive hyperemia and increases in wall shear rate that signal vasodilatory molecules to be released from the endothelium eliciting vasodilation. Despite the thousands of studies performing flow-mediated dilation in humans, surprisingly, no studies have performed this technique non-invasively in living rats. Considering the recent shift in focus to translational research, the establishment of guidelines for non-invasive measurement of flow-mediated dilation in rats and other rodents would be extremely valuable. In the following article, a protocol is presented for the non-invasive measurement of flow-mediated dilation in brachial and superficial femoral arteries of rats, as those sites are most commonly measured in humans.
Collapse
Affiliation(s)
| | - Miriam E Leary
- Department of Kinesiology and Health Education, University of Texas at Austin
| | - Yuxia He
- Department of Internal Medicine, University of Utah; Division of Nephrology and Hypertension, University of Utah
| | - Yan-Ting Shiu
- Department of Internal Medicine, University of Utah; Division of Nephrology and Hypertension, University of Utah
| | - Hirofumi Tanaka
- Department of Kinesiology and Health Education, University of Texas at Austin
| | - Anthony J Donato
- Department of Internal Medicine, University of Utah; Department of Biochemistry, University of Utah; Department of Exercise and Sport Science, University of Utah; Geriatric Research Education and Clinical Center, Department of Veterans Affairs
| |
Collapse
|
80
|
Kweider N, Huppertz B, Rath W, Lambertz J, Caspers R, ElMoursi M, Pecks U, Kadyrov M, Fragoulis A, Pufe T, Wruck CJ. The effects of Nrf2 deletion on placental morphology and exchange capacity in the mouse. J Matern Fetal Neonatal Med 2016; 30:2068-2073. [PMID: 27633272 DOI: 10.1080/14767058.2016.1236251] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Intrauterine growth restriction (IUGR) is defined as a pathological decreased fetal growth. Oxidative stress has been connected to the restriction in the fetal growth. The transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2) is a potent activator of the cellular antioxidant response. The effect Nrf2 on fetal-placental development has not yet been sufficiently investigated. Here, we evaluated the placental and fetal growth in Nrf2 knockout (Nrf2-KO) and Nrf2-wild type mice (Nrf2-WT) throughout pregnancy. METHODS Heterozygote Nrf2 (Nrf2+/-) mice were paired to get Nrf2-KO and Nrf2-WT in the litters. Placentae and embryos from both genotypes were collected and weighed on days 13.5, 15.5 and 18.5 post coitum. The absolute volumes of the labyrinth zone and the total volume of the placenta were determined using the Cavalieri principle. RESULTS On E 18.5 the fetal weight in Nrf2-KO was significantly reduced versus Nrf2-WT indicating a decrease in placental efficiency. A significant reduction in both total and labyrinth-volume in the placenta of Nrf2-KO mice was observed. CONCLUSION This data points out the necessity of functional Nrf2 for fetal and placental growth. A deficiency in Nrf2 signaling may negatively affect nutrient transfer capacity which is then no longer able to meet fetal growth demands.
Collapse
Affiliation(s)
- Nisreen Kweider
- a Department of Anatomy and Cell Biology , RWTH Aachen University Hospital , Aachen , Germany
| | - Berthold Huppertz
- b Institute of Cell Biology, Histology & Embryology, Medical University of Graz , Graz , Austria
| | - Werner Rath
- c Faculty of Medicine , Gynecology and Obstetrics, RWTH Aachen University Hospital , Aachen , Germany
| | - Jessica Lambertz
- d Institut of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital , Aachen , Germany
| | - Rebecca Caspers
- e Department of Obstetrics and Gynecology , RWTH Aachen University Hospital , Aachen , Germany
| | - Mohamed ElMoursi
- f Section of Obstetrics and Gynecology, Leeds Institute of Biomedical and Clinical sciences, University of Leeds , Leeds , UK.,g Department of Obstetrics and Gynecology , Mansoura University Faculty of Medicine , Mansoura , Egypt
| | - Ulrich Pecks
- e Department of Obstetrics and Gynecology , RWTH Aachen University Hospital , Aachen , Germany.,h Department of Obstetrics and Gynecology , University Hospital Schleswig-Holstein , Kiel , Germany , and
| | - Mamed Kadyrov
- a Department of Anatomy and Cell Biology , RWTH Aachen University Hospital , Aachen , Germany.,i Department of Neurology Mittelbaden Klinikum Baden-Baden , Baden-Baden , Germany
| | - Athanassios Fragoulis
- a Department of Anatomy and Cell Biology , RWTH Aachen University Hospital , Aachen , Germany
| | - Thomas Pufe
- a Department of Anatomy and Cell Biology , RWTH Aachen University Hospital , Aachen , Germany
| | - Christoph Jan Wruck
- a Department of Anatomy and Cell Biology , RWTH Aachen University Hospital , Aachen , Germany
| |
Collapse
|
81
|
Cortese-Krott MM, Pullmann D, Feelisch M. Nitrosopersulfide (SSNO -) targets the Keap-1/Nrf2 redox system. Pharmacol Res 2016; 113:490-499. [PMID: 27663261 DOI: 10.1016/j.phrs.2016.09.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/19/2016] [Accepted: 09/19/2016] [Indexed: 11/27/2022]
Abstract
Nitric oxide (NO), hydrogen sulfide and polysulfides have been proposed to contribute to redox signaling by activating the Keap-1/Nrf2 stress response system. Nitrosopersulfide (SSNO-) recently emerged as a bioactive product of the chemical interaction of NO or nitrosothiols with sulfide; upon decomposition it generates polysulfides and free NO, triggering the activation of soluble guanylate cyclase, inducing blood vessel relaxation in vitro and lowering blood pressure in vivo. Whether SSNO- itself interacts with the Keap-1/Nrf2 system is unknown. We therefore sought to investigate the ability of SSNO- to activate Nrf2-dependent processes in human vascular endothelial cells, and to compare the pharmacological effects of SSNO- with those of its precursors NO and sulfide at multiple levels of target engagement. We here demonstrate that SSNO- strongly increases nuclear levels, binding activity and transactivation activity of Nrf2, thereby increasing mRNA expression of Hmox-1, the gene encoding for heme oxygenase 1, without adversely affecting cell viability. Under all conditions, SSNO- appeared to be more potent than its parent compounds, NO and sulfide. SSNO--induced Nrf2 transactivation activity was abrogated by either NO scavenging with cPTIO or inhibition of thiol sulfuration by high concentrations of cysteine, implying a role for both persulfides/polysulfides and NO in SSNO- mediated Nrf2 activation. Taken together, our studies demonstrate that the Keap-1/Nrf2 redox system is a biological target of SSNO-, enriching the portfolio of bioactivity of this vasoactive molecule to also engage in the regulation of redox signaling processes. The latter suggests a possible role as messenger and/or mediator in cellular sensing and adaptations processes.
Collapse
Affiliation(s)
- Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany.
| | - David Pullmann
- Cardiovascular Research Laboratory, Department of Cardiology, Pneumology and Vascular Medicine, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, United Kingdom.
| |
Collapse
|
82
|
Wu H, Kong L, Tan Y, Epstein PN, Zeng J, Gu J, Liang G, Kong M, Chen X, Miao L, Cai L. C66 ameliorates diabetic nephropathy in mice by both upregulating NRF2 function via increase in miR-200a and inhibiting miR-21. Diabetologia 2016; 59:1558-1568. [PMID: 27115417 PMCID: PMC5260795 DOI: 10.1007/s00125-016-3958-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/23/2016] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Diabetic nephropathy is the leading cause of end-stage renal disease. Previously we reported that C66, a novel analogue of curcumin with a very high bioavailability, ameliorated diabetic nephropathy in mice, with little known about the mechanism. The present study aimed to define the mechanism by which C66 ameliorates diabetic nephropathy. METHODS Our aim was to discover whether C66 acts through the activation of nuclear factor (erythroid-derived 2)-like 2 (NFE2L2 or NRF2), which governs the antioxidant response. Streptozotocin-induced Nrf2 (also known as Nfe2l2)-knockout and wild-type (WT) diabetic mice were treated with C66. To determine whether the actions of C66 on NRF2 are mediated by microRNA (miR)-200a, WT diabetic mice were treated with C66 in the presence or absence of an in vivo miR-200a inhibitor (locked nucleic acid-modified anti-miR-200a [LNA-200a]) for 6 months. To determine whether miR-21 downregulation provided an NRF2-independent basis for C66 protection, Nrf2-knockout diabetic mice were treated with either C66 or an inhibitor of miR-21 (locked nucleic acid-modified anti-miR-21 [LNA-21]). RESULTS Deletion of Nrf2 partially abolished diabetic nephropathy protection by C66, confirming the requirement of NRF2 for this protection. Diabetic mice, but not C66-treated diabetic mice, developed significant albuminuria, renal oxidative damage and fibrosis. C66 upregulated renal miR-200a, inhibited kelch-like ECH-associated protein 1 and induced NRF2 function, effects that were prevented by LNA-200a. However, LNA-200a only partially reduced the protection afforded by C66, suggesting the existence of miR-200a/NRF2-independent mechanisms for C66 protection. C66 was also found to inhibit diabetes induction of miR-21. Both C66 and LNA-21 produced similar reductions in miR-21, albuminuria and renal fibrosis. CONCLUSIONS/INTERPRETATION The present study indicates that in addition to upregulating NRF2 by increasing miR-200a, C66 also protects against diabetic nephropathy by inhibiting miR-21.
Collapse
Affiliation(s)
- Hao Wu
- Department of Nephrology, the Second Hospital of Jilin University, 218 Ziqiang St, Changchun, Jilin, 130041, People's Republic of China
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Lili Kong
- Department of Nephrology, the First Hospital of Jilin University, Changchun, Jilin, People's Republic of China
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Yi Tan
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Paul N Epstein
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Jun Zeng
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Junlian Gu
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Maiying Kong
- Department of Bioinformatics and Biostatistics, SPHIS, University of Louisville, Louisville, KY, USA
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lining Miao
- Department of Nephrology, the Second Hospital of Jilin University, 218 Ziqiang St, Changchun, Jilin, 130041, People's Republic of China.
| | - Lu Cai
- Kosair Children's Hospital Research Institute at the Department of Pediatrics, Wendy L. Novak Diabetes Care Center, University of Louisville, 570 S Preston St, Baxter I., Louisville, KY, 40202, USA.
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
83
|
Clouet S, Di Pietrantonio L, Daskalopoulos EP, Esfahani H, Horckmans M, Vanorlé M, Lemaire A, Balligand JL, Beauloye C, Boeynaems JM, Communi D. Loss of Mouse P2Y6 Nucleotide Receptor Is Associated with Physiological Macrocardia and Amplified Pathological Cardiac Hypertrophy. J Biol Chem 2016; 291:15841-52. [PMID: 27231349 DOI: 10.1074/jbc.m115.684118] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Indexed: 12/13/2022] Open
Abstract
The study of the mechanisms leading to cardiac hypertrophy is essential to better understand cardiac development and regeneration. Pathological conditions such as ischemia or pressure overload can induce a release of extracellular nucleotides within the heart. We recently investigated the potential role of nucleotide P2Y receptors in cardiac development. We showed that adult P2Y4-null mice displayed microcardia resulting from defective cardiac angiogenesis. Here we show that loss of another P2Y subtype called P2Y6, a UDP receptor, was associated with a macrocardia phenotype and amplified pathological cardiac hypertrophy. Cardiomyocyte proliferation and size were increased in vivo in hearts of P2Y6-null neonates, resulting in enhanced postnatal heart growth. We then observed that loss of P2Y6 receptor enhanced pathological cardiac hypertrophy induced after isoproterenol injection. We identified an inhibitory effect of UDP on in vitro isoproterenol-induced cardiomyocyte hyperplasia and hypertrophy. The present study identifies mouse P2Y6 receptor as a regulator of cardiac development and cardiomyocyte function. P2Y6 receptor could constitute a therapeutic target to regulate cardiac hypertrophy.
Collapse
Affiliation(s)
- Sophie Clouet
- From the Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, 1070 Brussels
| | - Larissa Di Pietrantonio
- From the Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, 1070 Brussels
| | | | - Hrag Esfahani
- the Unit of Pharmacology and Therapeutics, Université Catholique de Louvain, UCL-FATH 5349, 1200 Brussels, and
| | - Michael Horckmans
- From the Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, 1070 Brussels
| | - Marion Vanorlé
- From the Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, 1070 Brussels
| | - Anne Lemaire
- From the Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, 1070 Brussels
| | - Jean-Luc Balligand
- the Unit of Pharmacology and Therapeutics, Université Catholique de Louvain, UCL-FATH 5349, 1200 Brussels, and
| | - Christophe Beauloye
- the Pole of Cardiovascular Research, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, 1200 Brussels
| | - Jean-Marie Boeynaems
- From the Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, 1070 Brussels, the Department of Laboratory Medicine, Erasme Hospital, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Didier Communi
- From the Institute of Interdisciplinary Research, IRIBHM, Université Libre de Bruxelles, 1070 Brussels,
| |
Collapse
|