1
|
Boothe PF, Kumar VP, Kong Y, Wang K, Levinson H, Mu D, Brown ML. Radiation Induced Skin Fibrosis (RISF): Opportunity for Angiotensin II-Dependent Intervention. Int J Mol Sci 2024; 25:8261. [PMID: 39125831 PMCID: PMC11312688 DOI: 10.3390/ijms25158261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Medical procedures, such as radiation therapy, are a vital element in treating many cancers, significantly contributing to improved survival rates. However, a common long-term complication of such exposure is radiation-induced skin fibrosis (RISF), a complex condition that poses substantial physical and psychological challenges. Notably, about 50% of patients undergoing radiation therapy may achieve long-term remission, resulting in a significant number of survivors managing the aftereffects of their treatment. This article delves into the intricate relationship between RISF, reactive oxygen species (ROS), and angiotensin II (Ang II) signaling. It proposes the underlying mechanisms and examines potential treatments for mitigating skin fibrosis. The primary goal is to offer essential insights in order to better care for and improve the quality of life of cancer survivors who face the risk of developing RISF.
Collapse
Affiliation(s)
- Patricia F. Boothe
- Department of Internal Medicine, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Vidya P. Kumar
- Armed Forces Radiobiology Research Institute, The Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Yali Kong
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (Y.K.); (D.M.)
| | - Kan Wang
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (Y.K.); (D.M.)
| | - Howard Levinson
- The Center for Plastic Surgery at Sentara, 301 Riverview Ave. #400, Norfolk, VA 23510, USA;
| | - David Mu
- Department of Biomedical and Translational Sciences, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; (Y.K.); (D.M.)
- Leroy T. Canoles Jr. Cancer Research Center, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Milton L. Brown
- Department of Internal Medicine, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| |
Collapse
|
2
|
Xu S, Han X, Wang X, Yu Y, Qu C, Liu X, Yang B. The role of oxidative stress in aortic dissection: a potential therapeutic target. Front Cardiovasc Med 2024; 11:1410477. [PMID: 39070552 PMCID: PMC11272543 DOI: 10.3389/fcvm.2024.1410477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
The incidence of aortic dissection (AD) is steadily increasing, driven by the rising prevalence of chronic conditions such as hypertension and the global aging of the population. Oxidative stress emerges as a pivotal pathophysiological mechanism contributing to the progression of AD. Oxidative stress triggers apoptosis in vascular smooth muscle cells, reshapes the extracellular matrix (ECM), and governs ECM degradation and remodeling, subsequently impacting aortic compliance. Furthermore, oxidative stress not only facilitates the infiltration of macrophages and mononuclear lymphocytes but also disrupts the integral structure and functionality of endothelial cells, thereby inducing endothelial cell dysfunction and furthering the degeneration of the middle layer of the aortic wall. Investigating antioxidants holds promise as a therapeutic avenue for addressing AD.
Collapse
Affiliation(s)
- Shengnan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Yi Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Khir NAM, Noh ASM, Long I, Zakaria R, Ismail CAN. Recent progress on anti-nociceptive effects of carbon monoxide releasing molecule-2 (CORM-2). Mol Cell Biochem 2024; 479:539-552. [PMID: 37106243 DOI: 10.1007/s11010-023-04749-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
The role of carbon monoxide (CO) has evolved albeit controversial disputes on its toxicity. This biological gasotransmitter participates in the endogenous regulation of neurotransmitters and neuropeptides released in the nervous system. Exogenous CO gas inhalation at a lower concentration has been the subject of investigations, which have revealed its biological homeostatic mechanisms and protective effects against many pathological conditions. This therapeutic procedure of CO is, however, limited due to its immediate release, which favours haemoglobin at a high affinity with the subsequent generation of toxic carboxyhaemoglobin in tissues. In order to address this problem, carbon monoxide releasing molecule-2 (CORM-2) or also known as tricarbonyldichlororuthenium II dimer is developed to liberate a controlled amount of CO in the biological systems. In this review, we examine several potential mechanisms exerted by this therapeutic compound to produce the anti-nociceptive effect that has been demonstrated in previous studies. This review could shed light on the role of CORM-2 to reduce pain, especially in cases of chronic and neuropathic pain.
Collapse
Affiliation(s)
- Nurul Ajilah Mohamed Khir
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
- International Medical School, Management and Science University, 40100, Shah Alam, Selangor, Malaysia
| | - Ain' Sabreena Mohd Noh
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Idris Long
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Rahimah Zakaria
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia
| | - Che Aishah Nazariah Ismail
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia.
- Brain and Behaviour Cluster, School of Medical Sciences, Universiti Sains Malaysia Health Campus, 16150, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
4
|
Shen D, Wu C, Fan G, Li T, Dou J, Zhu J, Li C, Kou X. Jujube peel polyphenols synergistically inhibit lipopolysaccharide-induced inflammation through multiple signaling pathways in RAW 264.7 cells. Food Chem Toxicol 2022; 164:113062. [PMID: 35460827 DOI: 10.1016/j.fct.2022.113062] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 12/11/2022]
Abstract
Jujube has great potential as food and traditional drugs in several countries. To study the anti-inflammatory influence of jujube peel polyphenols in lipopolysaccharide (LPS) induced RAW 264.7 cells through mitogen-activated protein kinase (MAPK), nuclear factor-kappa B (NF-κB) and nuclear erythroid 2-related factor 2 (Nrf2) signaling pathways. In this study, the phenolic composition of polyphenols in jujube peel was analyzed using LC-MS/MS, and which was confirmed that the main polyphenols were p-coumaric acid, catechin and rutin. Meanwhile, jujube peel polyphenols attenuated the generation of TNF-α, IL-1β, IL-6, NO and PGE2 by inhibiting MAPK and NF-κB signaling pathways. Additionally, jujube peel polyphenol activate Nrf2 from the cytoplasm to the nucleus, regulate antioxidant enzymes and pro-inflammatory cytokines, and reduce oxidative stress and inflammatory responses. Results obtained from this study suggest that jujube peel polyphenols may alleviate oxidative stress and inflammation by inhibiting MAPK and NF-κB and activating Nrf2 signaling pathways. Furthermore, jujube peel polyphenols have a synergistic effect in the treatment of LPS-induced inflammatory in RAW 264.7 cells. In conclusion, this study not only reveals the mechanism by which jujube peel polyphenols inhibit LPS-induced inflammation in RAW 264.7 cells, but also provides guidance for the development of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Dongbei Shen
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, Jiangsu, 210037, PR China
| | - Caie Wu
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, Jiangsu, 210037, PR China; Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China.
| | - Gongjian Fan
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, Jiangsu, 210037, PR China; Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China
| | - Tingting Li
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, Jiangsu, 210037, PR China; Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China
| | - Jinfeng Dou
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, Jiangsu, 210037, PR China
| | - Jinpeng Zhu
- College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing, Jiangsu, 210037, PR China
| | - Chunmei Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Xiuying Kou
- Infinitus (China) Company Ltd. Guangzhou, Guangdong, 510663, PR China
| |
Collapse
|
5
|
Sun HJ, Wang ZC, Nie XW, Bian JS. Therapeutic potential of carbon monoxide in hypertension-induced vascular smooth muscle cell damage revisited: from physiology and pharmacology. Biochem Pharmacol 2022; 199:115008. [PMID: 35318039 DOI: 10.1016/j.bcp.2022.115008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 01/14/2023]
Abstract
As a chronic and progressive disorder, hypertension remains to be a serious public health problem around the world. Among the different types of hypertension, pulmonary arterial hypertension (PAH) is a devastating disease associated with pulmonary arteriole remodeling, right ventricular failure and death. The contemporary management of systemic hypertension and PAH has substantially grown since more therapeutic targets and/or agents have been developed. Evolving treatment strategies targeting the vascular remodeling lead to improving outcomes in patients with hypertension, nevertheless, significant advancement opportunities for developing better antihypertensive drugs remain. Carbon monoxide (CO), an active endogenous gasotransmitter along with hydrogen sulfide (H2S) and nitric oxide (NO), is primarily generated by heme oxygenase (HO). Cumulative evidence suggests that CO is considered as an important signaling molecule under both physiological and pathological conditions. Studies have shown that CO confers a number of biological and pharmacological properties, especially its involvement in the pathological process and treatment of hypertension-related vascular remodeling. This review will critically outline the roles of CO in hypertension-associated vascular remodeling and discuss the underlying mechanisms for the protective effects of CO against hypertension and vascular remodeling. In addition, we will propose the challenges and perspectives of CO in hypertensive vascular remodeling. It is expected that a comprehensive understanding of CO in the vasculature might be essential to translate CO to be a novel pharmacological agent for hypertension-induced vascular remodeling.
Collapse
Affiliation(s)
- Hai-Jian Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Zi-Chao Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Xiao-Wei Nie
- Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518055, China.
| | - Jin-Song Bian
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu 215000, China.
| |
Collapse
|
6
|
Zhang X, Zhang H, Yang X, Qin Q, Sun X, Hou Y, Chen D, Jia M, Su X, Chen Y. Angiotensin II upregulates endothelin receptors through the adenosine monophosphate-activated protein kinase/sirtuin 1 pathway in vascular smooth muscle cells. J Pharm Pharmacol 2021; 73:1652-1662. [PMID: 34570873 DOI: 10.1093/jpp/rgab137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 08/26/2021] [Indexed: 11/13/2022]
Abstract
OBJECTIVES This study was designed to test our hypothesis that angiotensin II (Ang II) upregulates endothelin (ET) receptors in vascular smooth muscle cells (VSMCs). METHODS Rat superior mesenteric artery (SMA) without endothelium was cultured in serum-free medium for 24 h in the presence of Ang II with or without metformin or nicotinamide. In vivo, rats were implanted subcutaneously with a mini-osmotic pump infusing AngII (500 ng/kg/min) for 4 weeks. The level of protein expression was determined using Western blotting. The contractile response to ET receptor agonists was studied using sensitive myography. Caudal artery blood pressure (BP) was measured using non-invasive tail-cuff plethysmography. KEY FINDINGS The results showed that Ang II significantly increased ET receptors and decreased phosphorylated-adenosine monophosphate-activated protein kinase α (p-AMPKα) in SMA. Furthermore, metformin significantly inhibited Ang II-upregulated ET receptors and upregulated Ang II-decreased sirtuin 1 (Sirt1). However, this effect was reversed by nicotinamide. Moreover, the in-vivo results showed that metformin not only inhibited Ang II-induced upregulation of ET receptors but also recovered Ang II-decreased p-AMPKα and Sirt1. In addition, metformin significantly inhibited Ang II-elevated BP. However, the effect was reversed by nicotinamide, except for p-AMPKα. CONCLUSIONS Ang II upregulated ET receptors in VSMCs to elevate BP by inhibiting AMPK, thereby inhibiting Sirt1.
Collapse
Affiliation(s)
- Xin Zhang
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Hongmei Zhang
- The First Affiliated Hospital of Xi'an Medical University, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Xinpu Yang
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Qiaohong Qin
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Xia Sun
- School of Basic and Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Ying Hou
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Di Chen
- School of Basic and Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Min Jia
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Xingli Su
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
- School of Basic and Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Yulong Chen
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
7
|
Wu Q, Luo F, Wang XL, Lin Q, Liu GQ. Angiotensin I-converting enzyme inhibitory peptide: an emerging candidate for vascular dysfunction therapy. Crit Rev Biotechnol 2021; 42:736-755. [PMID: 34634988 DOI: 10.1080/07388551.2021.1948816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Abnormal vasoconstriction, inflammation, and vascular remodeling can be promoted by angiotensin II (Ang II) in the renin-angiotensin system (RAS), leading to vascular dysfunction diseases such as hypertension and atherosclerosis. Researchers have recently focused on angiotensin I-converting enzyme inhibitory peptides (ACEIPs), that have desirable efficacy in vascular dysfunction therapy due to Ang II reduction by inhibiting ACE activity. Promising methods for the large-scale preparation of ACEIPs include selective enzymatic hydrolysis and microbial fermentation. Thus far, ACEIPs have been widely reported to be hydrolyzed from protein-rich sources, including animals, plants, and marine organisms, while many emerging microorganism-derived ACEIPs are theoretically biosynthesized through the nonribosomal peptide synthase (NRPS) pathway. Notably, vasodilatation, anti-inflammation, and vascular reconstruction reversal of ACEIPs are strongly correlated. However, the related molecular mechanisms underlying signal transduction regulation in vivo remain unclear. We provide a comprehensive update of the ACE-Ang II-G protein-coupled type 1 angiotensin receptor (AT1R) axis signaling and its functional significance for potential translation into therapeutic strategies, particularly targeting AT1R by ACEIPs, as well as specific related signaling pathways. Future studies are expected to verify the biosynthetic regulatory mechanism of ACEIPs via the NRPS pathway, the effect of gut microbiota metabolism on vascular dysfunction and rigorous studies of ACE-Ang II-AT1R signaling pathways mediated by ACEIPs in large animals and humans.
Collapse
Affiliation(s)
- Qiang Wu
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China.,College of Food and Chemical Engineering, Shaoyang University, Shaoyang, China
| | - Feijun Luo
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China.,College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Xiao-Ling Wang
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China
| | - Qinlu Lin
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China.,College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Gao-Qiang Liu
- International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology of Hunan Province, Central South University of Forestry and Technology, Changsha, China
| |
Collapse
|
8
|
Chen YC, Chen JH, Tsai CF, Wu CT, Wu MH, Chang PC, Yeh WL. Nicardipine Inhibits Breast Cancer Migration via Nrf2/HO-1 Axis and Matrix Metalloproteinase-9 Regulation. Front Pharmacol 2021; 12:710978. [PMID: 34483918 PMCID: PMC8414136 DOI: 10.3389/fphar.2021.710978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/07/2021] [Indexed: 11/22/2022] Open
Abstract
Background: Metastasis represents an advanced stage of cancers, and matrix metalloproteinases are critical regulators. Calcium signal is crucial for appropriate cell behaviors. The efficacy and effects of calcium channel blockers in treating cancers are individually differ from each other. Here, we attempt to investigate the effects of nicardipine, a FDA-approved calcium channel blocker, in advanced breast cancers. Methods: We analyzed the influence of nicardipine on the colony-forming ability of triple negative breast cancer cell lines. Using cell culture inserts, cell migration was also examined. The expression of regulatory proteins was evaluated by real-time PCR, Western blot, and ELISA. Results: We have confirmed that nicardipine inhibits the breast cancer cells migration and colony formation. In addition, we also revealed that nicardipine increases the Nrf2 and HO-1 expression. The inhibition of HO-1 abrogates nicardipine-reduced matrix metalloproteinase-9 expression. Moreover, the end products of HO-1, namely, CO, Fe2+, and biliverdin (will converted to bilirubin), also decreases the expression of matrix metalloproteinase-9. Conclusion: These findings suggest that nicardipine-mediated matrix metalloproteinase-9 reduction is regulated by Nrf2/HO-1 axis and its catalytic end products. Therefore, nicardipine may be a potential candidate for repurposing against advanced breast cancers.
Collapse
Affiliation(s)
- Yen-Chang Chen
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Cheng-Fang Tsai
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Chen-Teng Wu
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Miao-Hsiang Wu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Wei-Lan Yeh
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,Institute of New Drug Development, China Medical University, Taichung, Taiwan.,Department of Biochemistry, School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
9
|
Carbon Monoxide-Releasing Molecule-2 Ameliorates Particulate Matter-Induced Aorta Inflammation via Toll-Like Receptor/NADPH Oxidase/ROS/NF- κB/IL-6 Inhibition. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2855042. [PMID: 34336088 PMCID: PMC8292097 DOI: 10.1155/2021/2855042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022]
Abstract
Particulate matter (PM), a major air pollutant, may be associated with adverse cardiovascular effects. Reactive oxygen species- (ROS-) dependent proinflammatory cytokine production, such as interleukin-6 (IL-6), is a possible underlying mechanism. Carbon monoxide- (CO-) releasing molecule-2 (CORM-2) which liberates exogenous CO can exert many beneficial effects, particularly anti-inflammation and antioxidant effects. The purpose of this study was to explore the protective effects and underpinning mechanisms of CORM-2 on PM-induced aorta inflammation. Here, human aortic vascular smooth muscle cells (HASMCs) were utilized as in vitro models for the assessment of signaling pathways behind CORM-2 activities against PM-induced inflammatory responses, including Toll-like receptors (TLRs), NADPH oxidase, ROS, nuclear factor-kappa B (NF-κB), and IL-6. The modulation of monocyte adherence and HASMC migration, that are two critical cellular events of inflammatory process, along with their regulators, including intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and matrix metalloproteinase-2 (MMP-2) and MMP-9, in response to PM by CORM-2, were further evaluated. Finally, mice experiments under different conditions were conducted for the in vivo evaluation of CORM-2 benefits on the expression of inflammatory molecules including IL-6, ICAM-1, VCAM-1, MMP-2, and MMP-9. Our results found that PM could induce aorta inflammation in vitro and in vivo, as evidenced by the increase of IL-6 expression that was regulated by the TLR2 and TLR4/NADPH oxidase/ROS/NF-κB signaling pathway, thereby promoting ICAM-1- and VCAM-1-dependent monocyte adhesion and MMP-2- and MMP-9-dependent HASMC migration. Importantly, our experimental models demonstrated that CORM-2-liberated CO effectively inhibited the whole identified PM-induced inflammatory cascade in HASMCs and tissues. In conclusion, CORM-2 treatment may elicit multiple beneficial effects on inflammatory responses of aorta due to PM exposure, thereby providing therapeutic value in the context of inflammatory diseases of the cardiovascular system.
Collapse
|
10
|
Ho CC, Chen YC, Tsai MH, Tsai HT, Weng CY, Yet SF, Lin P. Ambient Particulate Matter Induces Vascular Smooth Muscle Cell Phenotypic Changes via NOX1/ROS/NF-κB Dependent and Independent Pathways: Protective Effects of Polyphenols. Antioxidants (Basel) 2021; 10:antiox10050782. [PMID: 34069133 PMCID: PMC8156007 DOI: 10.3390/antiox10050782] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/06/2021] [Accepted: 05/13/2021] [Indexed: 11/16/2022] Open
Abstract
Epidemiological studies have demonstrated an association between ambient particulate matter (PM) exposure and vascular diseases. Here, we observed that treatment with ambient PM increased cell migration ability in vascular smooth muscle cells (VSMCs) and pulmonary arterial SMCs (PASMCs). These results suggest that VSMCs and PASMCs transitioned from a differentiated to a synthetic phenotype after PM exposure. Furthermore, treatment with PM increased intracellular reactive oxygen species (ROS), activated the NF-κB signaling pathway, and increased the expression of proinflammatory cytokines in VSMCs. Using specific inhibitors, we demonstrated that PM increased the migration ability of VSMCs via the nicotinamide–adenine dinucleotide phosphate (NADPH) oxidase 1 (NOX1)/ROS-dependent NF-κB signaling pathway, which also partially involved in the induction of proinflammatory cytokines. Finally, we investigated whether nature polyphenolic compounds prevent PM-induced migration and proinflammatory cytokines secretion in VSMCs. Curcumin, resveratrol, and gallic acid prevented PM2.5-induced migration via the ROS-dependent NF-κB signaling pathway. However, honokiol did not prevent PM2.5-induced migration or activation of the ROS-dependent NF-κB signaling pathway. On the other hand, all polyphenols prevented PM2.5-induced cytokines secretion. These data indicated that polyphenols prevented PM-induced migration and cytokine secretion via blocking the ROS-dependent NF-κB signaling pathway in VSMCs. However, other mechanisms may also contribute to PM-induced cytokine secretion.
Collapse
Affiliation(s)
- Chia-Chi Ho
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan 53053, Taiwan; (C.-C.H.); (Y.-C.C.); (M.-H.T.); (H.-T.T.); (C.-Y.W.)
| | - Yu-Cheng Chen
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan 53053, Taiwan; (C.-C.H.); (Y.-C.C.); (M.-H.T.); (H.-T.T.); (C.-Y.W.)
| | - Ming-Hsien Tsai
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan 53053, Taiwan; (C.-C.H.); (Y.-C.C.); (M.-H.T.); (H.-T.T.); (C.-Y.W.)
| | - Hui-Ti Tsai
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan 53053, Taiwan; (C.-C.H.); (Y.-C.C.); (M.-H.T.); (H.-T.T.); (C.-Y.W.)
| | - Chen-Yi Weng
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan 53053, Taiwan; (C.-C.H.); (Y.-C.C.); (M.-H.T.); (H.-T.T.); (C.-Y.W.)
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 53053, Taiwan
- Correspondence: (S.-F.Y.); (P.L.); Tel.: +886-37-246166 (ext. 38311) (S.-F.Y.); +886-37-246166 (ext. 36508) (P.L.)
| | - Pinpin Lin
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan 53053, Taiwan; (C.-C.H.); (Y.-C.C.); (M.-H.T.); (H.-T.T.); (C.-Y.W.)
- Correspondence: (S.-F.Y.); (P.L.); Tel.: +886-37-246166 (ext. 38311) (S.-F.Y.); +886-37-246166 (ext. 36508) (P.L.)
| |
Collapse
|
11
|
Que Y, Shu X, Wang L, Wang S, Li S, Hu P, Tong X. Inactivation of SERCA2 Cys 674 accelerates aortic aneurysms by suppressing PPARγ. Br J Pharmacol 2021; 178:2305-2323. [PMID: 33591571 DOI: 10.1111/bph.15411] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/24/2020] [Accepted: 02/04/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Inactivation of Cys674 (C674) in the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) causes intracellular Ca2+ accumulation, which activates calcineurin-mediated nuclear factor of activated T-lymphocytes (NFAT)/NF-κB pathways, and results in the phenotypic modulation of smooth muscle cells (SMCs) to accelerate angiotensin II-induced aortic aneurysms. Our goal was to investigate the mechanism involved. EXPERIMENTAL APPROACH We used heterozygous SERCA2 C674S knock-in (SKI) mice, where half of C674 was substituted by serine, to mimic partial irreversible oxidation of C674. The aortas of SKI mice and their littermate wild-type mice were collected for RNA sequencing, cell culture, protein expression, luciferase activity and aortic aneurysm analysis. KEY RESULTS Inactivation of C674 inhibited the promoter activity and protein expression of PPARγ, which could be reversed by inhibitors of calcineurin or NF-κB. In SKI SMCs, inhibition of NF-κB by pyrrolidinedithiocarbamic acid (PDTC) or overexpression of PPARγ2 reversed the protein expression of SMC phenotypic modulation markers and inhibited cell proliferation, migration, and macrophage adhesion to SMCs. Pioglitazone, a PPARγ agonist, blocked the activation of NFAT/NF-κB, reversed the protein expression of SMC phenotypic modulation markers, and inhibited cell proliferation, migration, and macrophage adhesion to SMCs in SKI SMCs. Furthermore, pioglitazone also ameliorated angiotensin II-induced aortic aneurysms in SKI mice. CONCLUSIONS AND IMPLICATIONS The inactivation of SERCA2 C674 promotes the development of aortic aneurysms by disrupting the balance between PPARγ and NFAT/NF-κB. Our study highlights the importance of C674 redox status in regulating PPARγ to maintain aortic homeostasis.
Collapse
Affiliation(s)
- Yumei Que
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xi Shu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Langtao Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Sai Wang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Siqi Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Pingping Hu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiaoyong Tong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
12
|
Huang KC, Li JC, Wang SM, Cheng CH, Yeh CH, Lin LS, Chiu HY, Chang CY, Chuu JJ. The effects of carbon monoxide releasing molecules on paraquat-induced pulmonary interstitial inflammation and fibrosis. Toxicology 2021; 456:152750. [PMID: 33737140 DOI: 10.1016/j.tox.2021.152750] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/10/2021] [Accepted: 03/12/2021] [Indexed: 01/07/2023]
Abstract
Paraquat, an herbicide used extensively worldwide, can cause severe toxicity in humans and animals, leading to irreversible, lethal lung fibrosis. The potential of CO-releasing molecules (CORMs), substances that release CO (Carbon monoxide) within animal tissues, for treating paraquat-induced ROS generation and inflammation is investigated here. Our results show that the fast CO releaser CORM-3 (4-20 μM) acts as a potential scavenger of free radicals and decreases fibrosis progression by inhibiting paraquat-induced overexpression of connective tissue growth factor and angiotensin II in MRC-5 cells. The slow CO releaser CORM-A1 (5 mg/kg) clearly decreased expression of the lung profibrogenic cytokines COX-2, TNF-α, and α-SMA and serum hydroxyproline, resulting in a lower mortality rate in paraquat-treated mice. Mice treated with higher-dose CORM-A1 (10 mg/kg) had relatively intact lung lobes and fewer fibrotic patches by gross observation, with less collagen deposition, mesangial matrix accumulation, and pulmonary fibrosis resulting from the mitigation of TGF-β overexpression. In conclusion, our data demonstrate for the first time that CORM-A1 alleviated the development of the fibrotic process and improved survival rate in mice exposed to PQ, would be an attractive therapeutic approach to attenuate the progression of pulmonary fibrosis following PQ exposure.
Collapse
Affiliation(s)
- Kuo-Ching Huang
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Hospital, Liouying, Tainan, Taiwan; Department of Environmental and Occupational Health, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Jui-Chen Li
- Pharmacy Department, Wei-Gong Memorial Hospital, Miaoli, Taiwan
| | - Shu-Mei Wang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei, Taiwan
| | - Chia-Hui Cheng
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chun-Hsiang Yeh
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Li-Syun Lin
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Hsin-Yi Chiu
- Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chia-Yu Chang
- Department of Neurology, Chi-Mei Medical Center, Tainan, Taiwan; Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| | - Jiunn-Jye Chuu
- Pharmacy Department, Wei-Gong Memorial Hospital, Miaoli, Taiwan; Department of Biotechnology and Food Technology, College of Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| |
Collapse
|
13
|
Targeting Heme Oxygenase-1 in the Arterial Response to Injury and Disease. Antioxidants (Basel) 2020; 9:antiox9090829. [PMID: 32899732 PMCID: PMC7554957 DOI: 10.3390/antiox9090829] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023] Open
Abstract
Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. The activation of vascular smooth muscle cells (SMCs) plays a critical role in mediating the aberrant arterial response to injury and a number of vascular diseases. Pharmacological induction or gene transfer of HO-1 improves arterial remodeling in animal models of post-angioplasty restenosis, vascular access failure, atherosclerosis, transplant arteriosclerosis, vein grafting, and pulmonary arterial hypertension, whereas genetic loss of HO-1 exacerbates the remodeling response. The vasoprotection evoked by HO-1 is largely ascribed to the generation of CO and/or the bile pigments, biliverdin and bilirubin, which exert potent antioxidant and anti-inflammatory effects. In addition, these molecules inhibit vascular SMC proliferation, migration, apoptosis, and phenotypic switching. Several therapeutic strategies are currently being pursued that may allow for the targeting of HO-1 in arterial remodeling in various pathologies, including the use of gene delivery approaches, the development of novel inducers of the enzyme, and the administration of unique formulations of CO and bilirubin.
Collapse
|
14
|
Yu W, Xiao L, Que Y, Li S, Chen L, Hu P, Xiong R, Seta F, Chen H, Tong X. Smooth muscle NADPH oxidase 4 promotes angiotensin II-induced aortic aneurysm and atherosclerosis by regulating osteopontin. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165912. [PMID: 32777344 DOI: 10.1016/j.bbadis.2020.165912] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Angiotensin II (Ang II) is commonly used to induce aortic aneurysm and atherosclerosis in animal models. Ang II upregulates NADPH oxidase isoform Nox4 in aortic smooth muscle cells (SMCs) in mice. However, whether smooth muscle Nox4 is directly involved in Ang II-induced aortic aneurysm and atherosclerosis is unclear. METHODS & RESULTS To address this, we used smooth muscle-specific Nox4 dominant-negative (SDN) transgenic mice, in which Nox4 activity is constitutively inhibited. In non-transgenic (NTg) mice, Ang II increased the expression of proteins known to contribute to both aortic aneurysm and atherosclerosis, namely osteopontin (OPN), collagen type I&III (Col I&III), matrix metalloproteinase 2 (MMP2), and vascular cell adhesion molecule 1 (VCAM1), which were all significantly downregulated in SDN mice. The number and size of Ang II-induced aorta collateral aneurysms and atherosclerotic lesions in the renal artery and aortic root of SDN mice were significantly decreased compared to NTg mice, and directly correlated with a decrease in OPN expression. Replenishing OPN in SDN SMCs, increased the expression of Col I&III, MMP2, and VCAM1, and promoted SMC proliferation, migration, and inflammation. CONCLUSIONS Our data demonstrate that smooth muscle Nox4 directly promotes the development of Ang II-induced aortic aneurysm and atherosclerosis, at least in part, through regulating OPN expression.
Collapse
Affiliation(s)
- Weimin Yu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Li Xiao
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yumei Que
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Siqi Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Lili Chen
- Wuhan EasyDiagnosis Biomedicine Co., Ltd., Wuhan 430075, China
| | - Pingping Hu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Rui Xiong
- Chongqing General Hospital, University of Chinese Academy of Science, Chongqing 400013, China
| | - Francesca Seta
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hao Chen
- Chongqing General Hospital, University of Chinese Academy of Science, Chongqing 400013, China
| | - Xiaoyong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
15
|
Juszczak M, Kluska M, Wysokiński D, Woźniak K. DNA damage and antioxidant properties of CORM-2 in normal and cancer cells. Sci Rep 2020; 10:12200. [PMID: 32699258 PMCID: PMC7376213 DOI: 10.1038/s41598-020-68948-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/30/2020] [Indexed: 12/21/2022] Open
Abstract
In this study, we compared the effect of tricarbonyldichlororuthenium (II) dimer (CORM-2) and its CO-depleted molecule (iCORM-2) on human peripheral blood mononuclear cells (PBMCs) and human promyelocytic leukemia HL-60 cells. We determined cell viability, DNA damage and DNA repair kinetics. We also studied the effect of both compounds on DNA oxidative damage, free radical level and HO-1 gene expression. We showed that at low concentrations both CORM-2 and iCORM-2 stimulate PBMCs viability. After 24-h incubation, CORM-2 and iCORM-2, at the concentration of 100 µM, reduce the viability of both PBMCs and HL-60 cells. We also demonstrated that CORM-2 and iCORM-2, in the 0.01–100 µM concentration range, cause DNA damage such as strand breaks and alkaline labile sites. DNA damage was repaired efficiently only in HL-60 cells. CORM-2 significantly reduces oxidative stress induced by 1 mM H2O2 in normal and cancer cells. On the contrary, iCORM-2 in HL-60 cells increases the level of free radicals in the presence of 1 and 5 mM H2O2. We also revealed that both CORM-2 and iCORM-2 induce HO-1 gene expression. However, CORM-2 induces this gene to a greater extent than iCORM-2, especially in HL-60 cells at 100 µM. Finally, we showed that CORM-2 and iCORM-2 reduce H2O2-induced DNA oxidative damage. Furthermore, CORM-2 proved to be a compound with stronger antioxidant properties than iCORM-2. Our results suggest that both active CORM-2 and inactive iCORM-2 exert biological effects such as cyto- and genotoxicity, antioxidant properties and the ability to induce the HO-1 gene. The released CO as well as iCORM-2 can be responsible for these effects.
Collapse
Affiliation(s)
- Michał Juszczak
- Faculty of Biology and Environmental Protection, Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Magdalena Kluska
- Faculty of Biology and Environmental Protection, Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Daniel Wysokiński
- Faculty of Biology and Environmental Protection, Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Katarzyna Woźniak
- Faculty of Biology and Environmental Protection, Department of Molecular Genetics, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland.
| |
Collapse
|
16
|
Li C, Zhong X, Xia W, He J, Gan H, Zhao H, Xia Y. The CX3CL1/CX3CR1 axis is upregulated in chronic kidney disease and contributes to angiotensin II-induced migration of vascular smooth muscle cells. Microvasc Res 2020; 132:104037. [PMID: 32615135 DOI: 10.1016/j.mvr.2020.104037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/10/2020] [Accepted: 06/23/2020] [Indexed: 01/20/2023]
Abstract
BACKGROUND The role of the chemokine axis, CX3CL1/CX3CR1, in the development of cardiovascular diseases has been widely speculated. Angiotensin II (Ang II) is a pivotal factor promoting cardiovascular complications in patients with chronic kidney disease (CKD). Whether there is a link between the two in CKD remains unclear. METHODS The uremic mice were treated with losartan for 8 weeks, and the expression of aortic CX3CL1/CX3CR1 was detected. Cultured mouse aortic vascular smooth muscle cells (VSMCs) were stimulated with Ang II, and then CX3CR1 expression was assessed by western blot. After the targeted disruption of CX3CR1 by transfection with siRNA, the migration of VSMCs was detected by transwell assay. Finally, both the activation of Akt pathway and the expression of IL-6 were detected by western blot. RESULTS Losartan treatment reduced the upregulation of aortic CX3CL1/CX3CR1 expression in uremic mice. In vitro, Ang II significantly upregulated CX3CR1 expression in VSMCs. Targeted disruption of CX3CR1 attenuated Ang II-induced migration of VSMCs. In addition, the use of CX3CR1-siRNA suppressed Akt phosphorylation and IL-6 production in VSMCs stimulated by Ang II. CONCLUSIONS The aortic CX3CL1/CX3CR1 is upregulated by Ang II in CKD, and it contributes to Ang II-induced migration of VSMCs in vitro.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Aorta/pathology
- CX3C Chemokine Receptor 1/genetics
- CX3C Chemokine Receptor 1/metabolism
- Cell Line
- Cell Movement/drug effects
- Chemokine CX3CL1/genetics
- Chemokine CX3CL1/metabolism
- Disease Models, Animal
- Interleukin-6/metabolism
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Signal Transduction
- Up-Regulation
- Uremia/metabolism
- Uremia/pathology
Collapse
Affiliation(s)
- Chengsheng Li
- Department of General Internal Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoyi Zhong
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wenyu Xia
- Class 4, Grade 2, Guangzhou Zhixin High School, Guangzhou 511430, China
| | - Jin He
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Hua Gan
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - HongFei Zhao
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Yunfeng Xia
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
17
|
Zhao XL, Yu L, Zhang SD, Ping K, Ni HY, Qin XY, Zhao CJ, Wang W, Efferth T, Fu YJ. Cryptochlorogenic acid attenuates LPS-induced inflammatory response and oxidative stress via upregulation of the Nrf2/HO-1 signaling pathway in RAW 264.7 macrophages. Int Immunopharmacol 2020; 83:106436. [PMID: 32234671 DOI: 10.1016/j.intimp.2020.106436] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/19/2020] [Accepted: 03/19/2020] [Indexed: 02/08/2023]
Abstract
Phenolic acids are found in natural plants, such as caffeic acid, rosmarinic acid, and chlorogenic acid. They have long been used as pharmacological actives, owing to their anti-inflammatory and antioxidant activities. Cryptochlorogenic acid (CCGA) is a special isomer of chlorogenic acid; the pharmacological effects and related molecular mechanisms of CCGA have been poorly reported. In the present study, the antioxidant and anti-inflammatory effects of CCGA in RAW 264.7 macrophages and the underlying mechanisms were investigated. The results revealed that CCGA dose-dependently inhibited LPS-induced production of NO, TNF-α, and IL-6 and blocked iNOS, COX-2, TNF-α, and IL-6 expressions. CCGA also significantly increased the GSH/GSSG ratio and SOD activity and reduced the MDA level. Moreover, CCGA suppressed the nuclear translocation of NF-κB by hindering the phosphorylation of IκB kinase (IKK) and degrading IκB. It also downregulated the phosphorylation of MAPKs. Our results indicated that CCGA significantly inhibited NF-κB activation by controlling the expression of pro-inflammatory factors and promoting the nuclear transfer of Nrf2. In conclusion, CCGA could attenuate LPS-induced inflammatory symptoms by modulating NF-κB/MAPK signaling cascades and inhibit LPS-induced oxidative stress via Nrf2 nuclear translocation.
Collapse
Affiliation(s)
- Xue-Lian Zhao
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Liang Yu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Sun-Dong Zhang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Kou Ping
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Hai-Yan Ni
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Xiang-Yu Qin
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Chun-Jian Zhao
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Wei Wang
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Thomas Efferth
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Yu-Jie Fu
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, China; Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; College of Forestry, Beijing Forestry University, Beijing 100083, China; Beijing Advanced Innovation Center for Tree Breeding by Molecular Design, Beijing Forestry University, Beijing 100083, China.
| |
Collapse
|
18
|
Yan Y, Wang L, Chen S, Zhao G, Fu C, Xu B, Tan X, Xiang Y, Chen G. Carbon Monoxide Inhibits T Cell Proliferation by Suppressing Reactive Oxygen Species Signaling. Antioxid Redox Signal 2020; 32:429-446. [PMID: 31810391 DOI: 10.1089/ars.2019.7814] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aims: Carbon monoxide (CO) confers antiproliferative effects on T cells; however, how these effects are produced remains unclear. Reactive oxygen species (ROS) have recently emerged as important modulators of T cell proliferation. In this study, we aimed to determine whether the inhibitory effects of CO on T cell proliferation are dependent on the inhibition of ROS signaling. Results: Pretreatment with CO-releasing molecule-2 (CORM-2) had potent inhibitory effects on mouse T cell proliferation stimulated by anti-CD3/CD28 antibodies. Interestingly, CORM-2 pretreatment markedly suppressed intracellular ROS generation as well as the activity of NADPH oxidase and mitochondrial complexes I-IV in T cells after stimulation. The inhibitory effects of CORM-2 on both ROS production and T cell proliferation were comparable with those produced by the use of antioxidant N-acetylcysteine or a combined administration of mitochondrial complex I-IV inhibitors. Moreover, increasing intracellular ROS via hydrogen peroxide supplementation largely reversed the inhibitory effect of CORM-2 on the proliferation of T cells. The inhibitory effects of CORM-2 on both cell proliferation and intracellular ROS production were also shown in a T cell proliferation model involving stimulation by allogeneic dendritic cells or phorbol 12-myristate 13-actetate/ionomycin, as well as in spontaneous cell proliferation models in EL-4 and RAW264.7 cells. In addition, CORM-2 treatment significantly inhibited T cell activation in vivo and attenuated concanavalin A-induced autoimmune hepatitis. Innovation: CO inhibits T cell proliferation via suppression of intracellular ROS production. Conclusion: The study could supply a general mechanism to explain the inhibitory effects of CO on T cell activation and proliferation, favoring its future application in T cell-mediated diseases.
Collapse
Affiliation(s)
- Yutao Yan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lu Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Song Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Guangyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Cheng Fu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingyang Xu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaosheng Tan
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Ying Xiang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
19
|
Inactivation of cysteine 674 in the SERCA2 accelerates experimental aortic aneurysm. J Mol Cell Cardiol 2020; 139:213-224. [PMID: 32035136 DOI: 10.1016/j.yjmcc.2020.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/15/2020] [Accepted: 02/03/2020] [Indexed: 01/12/2023]
Abstract
Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) is vital to maintain intracellular calcium homeostasis. SERCA2 cysteine 674 (C674) is highly conservative and its irreversible oxidation is upregulated in human and mouse aortic aneurysms, especially in smooth muscle cells (SMCs). The contribution of SERCA2 and its redox C674 in the development of aortic aneurysm remains enigmatic. Objective: Our goal was to investigate the contribution of inactivation of C674 to the development of aortic aneurysm and the mechanisms involved. Approach and results: Using SERCA2 C674S knock-in (SKI) mouse line, in which half of C674 was substituted by serine 674 (S674) to represent partial irreversible oxidation of C674 in aortic aneurysm, we found that in aortic SMCs the replacement of C674 by S674 resulted in SMC phenotypic modulation. In SKI SMCs, the increased intracellular calcium activated calcium-dependent calcineurin, which promoted the nuclear translocation of nuclear factor of activated T-lymphocytes (NFAT) and nuclear factor kappa-B (NFκB), while inhibition of calcineurin blocked SMC phenotypic modulation. Besides, the replacement of C674 by S674 accelerated angiotensin II-induced aortic aneurysm. Conclusions: Our results indicate that the inactivation of C674 by causing the accumulation of intracellular calcium to activate calcineurin-mediated NFAT/NFκB pathways, resulted in SMC phenotypic modulation to accelerate aortic aneurysm, which highlights the importance of C674 redox state in the development of aortic aneurysms.
Collapse
|
20
|
Abstract
Inhalation of high concentrations of carbon monoxide (CO) is known to lead to serious systemic complications and neuronal disturbances. However, it has been found that not only is CO produced endogenously, but also that low concentrations can bestow beneficial effects which may be of interest in biology and medicine. As translocation of CO through the human organism is difficult, small molecules known as CO-releasing molecules (CORMs) deliver controlled amounts of CO to biological systems, and these are of great interest from a medical point of view. These actions may prevent vascular dysfunction, regulate blood pressure, inhibit blood platelet aggregation or have anti-inflammatory effects. This review summarizes the functions of various CO-releasing molecules in biology and medicine.
Collapse
|
21
|
Tsai CF, Chen JH, Chang CN, Lu DY, Chang PC, Wang SL, Yeh WL. Fisetin inhibits cell migration via inducing HO-1 and reducing MMPs expression in breast cancer cell lines. Food Chem Toxicol 2018; 120:528-535. [DOI: 10.1016/j.fct.2018.07.059] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 01/09/2023]
|
22
|
An Overview of the Potential Therapeutic Applications of CO-Releasing Molecules. Bioinorg Chem Appl 2018; 2018:8547364. [PMID: 30158958 PMCID: PMC6109489 DOI: 10.1155/2018/8547364] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/19/2018] [Accepted: 07/18/2018] [Indexed: 02/08/2023] Open
Abstract
Carbon monoxide (CO) has long been known as the “silent killer” owing to its ability to form carboxyhemoglobin—the main cause of CO poisoning in humans. Its role as an endogenous neurotransmitter, however, was suggested in the early 1990s. Since then, the biological activity of CO has been widely examined via both the direct administration of CO and in the form of so-called “carbon monoxide releasing molecules (CORMs).” This overview will explore the general physiological effects and potential therapeutic applications of CO when delivered in the form of CORMs.
Collapse
|
23
|
Carbon monoxide-releasing molecule-3 protects against ischemic stroke by suppressing neuroinflammation and alleviating blood-brain barrier disruption. J Neuroinflammation 2018; 15:188. [PMID: 29929562 PMCID: PMC6014004 DOI: 10.1186/s12974-018-1226-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 06/14/2018] [Indexed: 02/08/2023] Open
Abstract
Background At low levels, carbon monoxide (CO) has been shown to have beneficial effects on multiple organs and tissues through its potential anti-inflammatory, anti-apoptotic, and anti-proliferative properties. However, the effect of CO-releasing molecule (CORM)-3, a water-soluble CORM, on ischemic stroke and its mechanism of action are still unclear. Methods We investigated the role of CORM-3 in the mouse model of transient middle cerebral artery occlusion (tMCAO). CORM-3 or saline was administered to mice by retro-orbital injection at the time of reperfusion after 1-h tMCAO or at 1 h after sham surgery. We assessed infarct volume and brain water content at 24 and 72 h after ischemia, blood-brain barrier permeability at 6 and 72 h after ischemia, and neurologic deficits on days 1, 3, 7, and 14. Results Among mice that underwent tMCAO, those that received CORM-3 had significantly smaller infarct volume and greater expression of neuronal nuclear antigen (NeuN) and microtubule-associated protein 2 than did saline-treated mice. CORM-3-treated mice had significantly fewer activated microglia in the peri-infarction zone than did control mice and exhibited downregulated expression of ionized calcium-binding adapter molecule (Iba)-1, tumor necrosis factor-α, and interleukin 1β. CORM-3-treated mice had significantly lower brain water content and enhanced neurologic outcomes on days 3, 7, and 14 post-tMCAO. Lastly, CORM-3 treatment reduced Evans blue leakage; increased expression of platelet-derived growth factor receptor-β, tight junction protein ZO-1, and matrix protein laminin; and decreased protein level of matrix metalloproteinase-9. Conclusion CORM-3 treatment at the time of reperfusion reduces ischemia-reperfusion-induced brain injury by suppressing neuroinflammation and alleviating blood-brain barrier disruption. Our data suggest that CORM-3 may provide an effective therapy for ischemic stroke.
Collapse
|
24
|
Abstract
Advancing age will affect every individual and its impact on cardiac health deserves significant attention. The age-related physiological changes occurring in the coronary vasculature, myocardium, and valves set the stage upon which cardiovascular disease can escalate in the elderly population. The overall focus of this review is to highlight new and noteworthy studies and to incorporate reviews related to cardiac senescence in the context of the current state of the field. Lastly, future directions in the field of cardiac aging and the development of novel therapeutics to treat pathophysiological conditions typically associated with advancing age will be discussed.
Collapse
Affiliation(s)
- Amanda J LeBlanc
- Cardiovascular Innovation Institute, Department of Physiology, 302 E. Muhammad Ali Blvd, University of Louisville, Louisville, KY 40202
| | - Natia Q Kelm
- Cardiovascular Innovation Institute, Department of Physiology, 302 E. Muhammad Ali Blvd, University of Louisville, Louisville, KY 40202
| | - Monika George
- Cardiovascular Innovation Institute, Department of Physiology, 302 E. Muhammad Ali Blvd, University of Louisville, Louisville, KY 40202
| |
Collapse
|
25
|
Ma F, Yang Y, Wang JD, Quan ZW, Zhou D. Helicobacter pylori and 17β-estradiol induce human intrahepatic biliary epithelial cell abnormal proliferation and oxidative DNA damage. Hepatobiliary Pancreat Dis Int 2017; 16:519-527. [PMID: 28992885 DOI: 10.1016/s1499-3872(17)60038-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 06/23/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Biliary cancers are more common in females, and previous studies have suggested that Helicobacter pylori (H. pylori) exists in the biliary system. However, the effects of H. pylori infection and estrogen on the biological behaviors of human biliary epithelium mucosa remain unknown. The present study aimed to clarify their effects on the proliferation, apoptosis, migration and oxidative DNA damage of a human intrahepatic biliary epithelial cell (HIBEC) line in vitro. METHODS HIBECs were co-cultured with 17β-estradiol (at 10-9 mol/L, 10-7 mol/L, and 10-5 mol/L) and H. pylori (at MOI=0.5:1, 1:1, and 2:1) and continuously passaged until the 15th generation (approximately 45 days). Then, the following assays were performed. HIBEC proliferation was measured using the CCK-8 assay, plate clone-formation assay and by determining Ki-67 expression with immunocytochemistry; cell apoptosis and migration were investigated using Annexin-V/PI and transwell assays, respectively; and reactive oxygen species (ROS) and 8-hydroxy-2'-deoxyguanosine (8-OHdG) production were detected by flow cytometry and immunofluorescence staining combined with confocal laser scanning microscopy, respectively. The results were the basis for evaluating the level of oxidative stress and the related DNA damage in HIBECs. RESULTS HIBECs maintained a normal morphology and vitality when treated with 17β-estradiol (at 10-9 mol/L) and H. pylori (at MOI=0.5:1 and 1:1). 17β-estradiol at 10-7 mol/L and 10-5 mol/L and H. pylori at MOI=2:1, by contrast, caused cell death. Compared with controls, HIBECs treated with 17β-estradiol (10-9 mol/L) and H. pylori (MOI=1:1) had a higher up-regulation of proliferation, Ki-67 expression, clone formation, migration activity and the expression of ROS and 8-OHdG and exhibited a down-regulation of apoptosis. The above effects were further increased when 17β-estradiol and H. pylori were combined (P<0.05). CONCLUSIONS H. pylori and 17β-estradiol, separately or in combination, promoted cell proliferation and suppressed apoptosis of HIBECs in vitro. The above phenomena might be related to oxidative stress and its subsequent DNA damage with H. pylori and 17β-estradiol.
Collapse
Affiliation(s)
- Fei Ma
- Department of Oncology, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, China
| | - Yong Yang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, China
| | - Jian-Dong Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, China
| | - Zhi-Wei Quan
- Department of General Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, China
| | - Di Zhou
- Department of General Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, China.
| |
Collapse
|
26
|
Adach W, Olas B. The role of CORM-2 as a modulator of oxidative stress and hemostatic parameters of human plasma in vitro. PLoS One 2017; 12:e0184787. [PMID: 28950024 PMCID: PMC5614530 DOI: 10.1371/journal.pone.0184787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 08/30/2017] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The main aim of the experiment is to examine the effect of CORM-2, a donor of carbon monoxide (CO), on oxidative stress in human plasma in vitro. In addition, it examines the effects of CORM-2 on the hemostatic parameters of plasma: the activated partial thromboplastin time (APTT), thrombin time (TT) and prothrombin time (PT). METHODS Human plasma was incubated for 5-60 min with different concentrations of CORM-2: 0.1-100 μM. Following this, various hemostatic factors and biomarkers of oxidative stress were studied. Lipid peroxidation was measured as thiobarbituric acid reactive substance (TBARS) concentration, and the oxidation of amino acid residues in proteins was measured by determining the amounts of carbonyl and thiol groups. RESULTS Two oxidative stress inducers: hydrogen peroxide (H2O2) and the donor of hydroxyl radical (H2O2/Fe) were used. Decrease in protein carbonylation, thiol group oxidation and lipid peroxidation were detected at tested concentrations of CORM-2. CONCLUSION Our results indicate that CORM-2 may have antioxidant properties in human plasma treated with H2O2 or H2O2/Fe. In addition, our results indicate the anti-coagulant activities of CORM-2 in vitro.
Collapse
Affiliation(s)
- Weronika Adach
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
- * E-mail:
| |
Collapse
|
27
|
CO-releasing molecules-2 attenuates ox-LDL-induced injury in HUVECs by ameliorating mitochondrial function and inhibiting Wnt/β-catenin pathway. Biochem Biophys Res Commun 2017. [DOI: 10.1016/j.bbrc.2017.06.089] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|