51
|
Li W, Luo Y, Huang Z, Shen S, Dai C, Shen S, Qi X, Liang G, Luo W. Costunolide Protects Myocardium From Ischemia Reperfusion Injury by Inhibiting Oxidative Stress Through Nrf2/Keap1 Pathway Activation. J Cardiovasc Pharmacol 2023; 82:117-127. [PMID: 37000981 DOI: 10.1097/fjc.0000000000001422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 03/16/2023] [Indexed: 06/19/2023]
Abstract
ABSTRACT Costunolide (Cos) is a naturally occurring sesquiterpene lactone that exhibits antioxidative properties. In this study, we demonstrate the protective mechanism of Cos against ischemia/reperfusion (I/R)-induced myocardial injury. Cos significantly decreased levels of reactive oxygen species and ameliorated apoptosis of I/R cardiomyocytes both in vitro and in vivo. Further investigation revealed that Cos increased expression of the antioxidant proteins HO-1 and NQO-1 and decreased the Bax/Bcl-2 ratio, thus protecting cardiac cells. NF-E2-related factor 2 (Nrf2) silencing significantly attenuated the protective effects of Cos in tert-butyl hydroperoxide (TBHP)-treated H9C2 cells. Additionally, Cos significantly intensified the I/R- or TBHP-induced dissociation of the Kelch-like ECH-associated protein 1 (Keap1)/Nrf2 complex both in vitro and in vivo. These results suggest that activation of Nrf2/Keap1 using Cos may be a therapeutic strategy for myocardial I/R injury.
Collapse
Affiliation(s)
- Weixin Li
- Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yue Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuqi Huang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Siyuan Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengyi Dai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sirui Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoxiao Qi
- Department of Pharmacy, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; and
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wu Luo
- Medical Research Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
52
|
Norton CE, Shaw RL, Segal SS. Differential Effects of High Fat Diets on Resilience to H 2O 2-Induced Cell Death in Mouse Cerebral Arteries: Role for Processed Carbohydrates. Antioxidants (Basel) 2023; 12:1433. [PMID: 37507971 PMCID: PMC10376469 DOI: 10.3390/antiox12071433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
High fat, western-style diets increase vascular oxidative stress. We hypothesized that smooth muscle cells and endothelial cells adapt during the consumption of high fat diets to become more resilient to acute oxidative stress. Male C57Bl/6J mice were fed a western-style diet high in fat and processed carbohydrates (WD), a high fat diet that induces obesity (DIO), or their respective control (CD) and standard (SD) diets for 16 weeks. Posterior cerebral arteries (PCAs) were isolated and pressurized for study. During acute exposure to H2O2 (200 µM), smooth muscle cell and endothelial cell death were reduced in PCAs from WD, but not DIO mice. WD selectively attenuated mitochondrial membrane potential depolarization and vessel wall Ca2+ influx during H2O2 exposure. Selective inhibition of transient receptor potential (TRP) V4 or TRPC3 channels reduced smooth muscle cell and endothelial cell death in concert with the vessel wall [Ca2+]i response to H2O2 for PCAs from CD mice and eliminated differences between CD and WD. Inhibiting Src kinases reduced smooth muscle cell death along with [Ca2+]i response to H2O2 only in PCAs from CD mice and eliminated differences between diets. However, Src kinase inhibition did not alter endothelial cell death. These findings indicate that consuming a WD, but not high fat alone, leads to adaptations that limit Ca2+ influx and vascular cell death during exposure to acute oxidative stress.
Collapse
Affiliation(s)
- Charles E Norton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | - Rebecca L Shaw
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
- Dalton Cardiovascular Research Center, Columbia, MO 65211, USA
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65201, USA
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, MO 65211, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
53
|
Ullah K, Ai L, Humayun Z, Wu R. Targeting Endothelial HIF2α/ARNT Expression for Ischemic Heart Disease Therapy. BIOLOGY 2023; 12:995. [PMID: 37508425 PMCID: PMC10376750 DOI: 10.3390/biology12070995] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
Ischemic heart disease (IHD) is a major cause of mortality and morbidity worldwide, with novel therapeutic strategies urgently needed. Endothelial dysfunction is a hallmark of IHD, contributing to its development and progression. Hypoxia-inducible factors (HIFs) are transcription factors activated in response to low oxygen levels, playing crucial roles in various pathophysiological processes related to cardiovascular diseases. Among the HIF isoforms, HIF2α is predominantly expressed in cardiac vascular endothelial cells and has a key role in cardiovascular diseases. HIFβ, also known as ARNT, is the obligate binding partner of HIFα subunits and is necessary for HIFα's transcriptional activity. ARNT itself plays an essential role in the development of the cardiovascular system, regulating angiogenesis, limiting inflammatory cytokine production, and protecting against cardiomyopathy. This review provides an overview of the current understanding of HIF2α and ARNT signaling in endothelial cell function and dysfunction and their involvement in IHD pathogenesis. We highlight their roles in inflammation and maintaining the integrity of the endothelial barrier, as well as their potential as therapeutic targets for IHD.
Collapse
Affiliation(s)
- Karim Ullah
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| | - Lizhuo Ai
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
- The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zainab Humayun
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| | - Rongxue Wu
- Section of Cardiology, Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
54
|
Savovic Z, Pindovic B, Nikolic M, Simic I, Davidovic G, Ignjatovic V, Vuckovic J, Zornic N, Nikolic Turnic T, Zivkovic V, Srejovic I, Bolevich S, Jakovljevic V, Iric Cupic V. Prognostic Value of Redox Status Biomarkers in Patients Presenting with STEMI or Non-STEMI: A Prospective Case-Control Clinical Study. J Pers Med 2023; 13:1050. [PMID: 37511663 PMCID: PMC10381258 DOI: 10.3390/jpm13071050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/26/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: The aim of our study was to determine the role of oxidative stress (OS) during early evaluation of acute ST-elevated myocardial infarction (STEMI) and non-ST-elevated myocardial infarction (NSTEMI) patients in order to define the role of redox balance in profiling the development of myocardial infarction (MI). (2) Methods: This prospective observational case-control study included 40 consecutive STEMI and 39 NSTEMI patients hospitalized in the coronary care unit of the cardiology clinic at the Kragujevac Clinical Center, Serbia, between 1 January 2016 and 1 January 2017. Blood samples were collected from all patients for measuring cardio-specific enzymes at admission and 12 h after admission to evaluate systemic oxidative stress biomarkers and the activity of antioxidant enzymes. (3) Results: In this study, participants were predominately female (52%), with a mean age of 56.17 ± 1.22 years old in the STEMI group and 69.17 ± 3.65 in the non-STEMI group. According to the Killip classification, the majority of patients (>50%) were at the second and third level. We confirmed the elevation of superoxide anion radicals in the non-STEMI group 6 h after admission in comparison with the STEMI and CTRL groups, but levels had decreased 12 h after admission. Levels of hydrogen peroxide were statistically significantly increased in the NSTEMI group. A positive correlation of superoxide anion radicals and levels of troponin I at admission was observed (r = 0.955; p = 0.045), as well as an inverse correlation between reduced glutathione and levels of NT-pBNP measured 6 h after admission (r = -0.973; p = 0.027). (4) Conclusions: We confirmed that superoxide anion radicals and reduced glutathione observed together with hs-troponin I at admission and NT-pBNP during hospital treatment could be predictors of ST evolution.
Collapse
Affiliation(s)
- Zorica Savovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
| | - Bozidar Pindovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Maja Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivan Simic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Goran Davidovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Vladimir Ignjatovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Jelena Vuckovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| | - Nenad Zornic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Tamara Nikolic Turnic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- N.A. Semashko Public Health and Healthcare Department, F. F. Erismann Institute of Public Health, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
| | - Vladimir Zivkovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
- Department of Clinical Pharmacology, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Ivan Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
- Department of Clinical Pharmacology, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Sergej Bolevich
- Department of Human Pathology, 1st Moscow State Medical, University I. M. Sechenov, 119991 Moscow, Russia
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia
- Department of Human Pathology, 1st Moscow State Medical, University I. M. Sechenov, 119991 Moscow, Russia
| | - Violeta Iric Cupic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (Z.S.)
- Department of Cardiology, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
55
|
Yang A, Guo L, Zhang Y, Qiao C, Wang Y, Li J, Wang M, Xing J, Li F, Ji L, Guo H, Zhang R. MFN2-mediated mitochondrial fusion facilitates acute hypobaric hypoxia-induced cardiac dysfunction by increasing glucose catabolism and ROS production. Biochim Biophys Acta Gen Subj 2023:130413. [PMID: 37331409 DOI: 10.1016/j.bbagen.2023.130413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND Rapid ascent to high-altitude environment which is characterized by acute hypobaric hypoxia (HH) may increase the risk of cardiac dysfunction. However, the potential regulatory mechanisms and prevention strategies for acute HH-induced cardiac dysfunction have not been fully clarified. Mitofusin 2 (MFN2) is highly expressed in the heart and is involved in the regulation of mitochondrial fusion and cell metabolism. To date, however, the significance of MFN2 in the heart under acute HH has not been investigated. METHODS AND RESULTS Our study revealed that MFN2 upregulation in hearts of mice during acute HH led to cardiac dysfunction. In vitro experiments showed that the decrease in oxygen concentration induced upregulation of MFN2, impairing cardiomyocyte contractility and increasing the risk of QT prolongation. Additionally, acute HH-induced MFN2 upregulation promoted glucose catabolism and led to excessive mitochondrial reactive oxygen species (ROS) production in cardiomyocytes, ultimately resulting in decreased mitochondrial function. Furthermore, co-immunoprecipitation (co-IP) and mass spectrometry analyses indicated that MFN2 interacted with the NADH-ubiquinone oxidoreductase 23 kDa subunit (NDUFS8). Specifically, acute HH-induced MFN2 upregulation increased NDUFS8-dependent complex I activity. CONCLUSIONS Taken together, our studies provide the first direct evidence that MFN2 upregulation exacerbates acute HH-induced cardiac dysfunction by increasing glucose catabolism and ROS production. GENERAL SIGNIFICANCE Our studies indicate that MFN2 may be a promising therapeutic target for cardiac dysfunction under acute HH.
Collapse
Affiliation(s)
- Ailin Yang
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Lifei Guo
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yanfang Zhang
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Chenjin Qiao
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yijin Wang
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Jiaying Li
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Min Wang
- College of Life Sciences, Northwest University, Xi'an 710069, China; Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, China
| | - Fei Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Lele Ji
- Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Haitao Guo
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Ru Zhang
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
56
|
Lee SS, Vũ TT, Weiss AS, Yeo GC. Stress-induced senescence in mesenchymal stem cells: Triggers, hallmarks, and current rejuvenation approaches. Eur J Cell Biol 2023; 102:151331. [PMID: 37311287 DOI: 10.1016/j.ejcb.2023.151331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as promising cell-based therapies in the treatment of degenerative and inflammatory conditions. However, despite accumulating evidence of the breadth of MSC functional potency, their broad clinical translation is hampered by inconsistencies in therapeutic efficacy, which is at least partly due to the phenotypic and functional heterogeneity of MSC populations as they progress towards senescence in vitro. MSC senescence, a natural response to aging and stress, gives rise to altered cellular responses and functional decline. This review describes the key regenerative properties of MSCs; summarises the main triggers, mechanisms, and consequences of MSC senescence; and discusses current cellular and extracellular strategies to delay the onset or progression of senescence, or to rejuvenate biological functions lost to senescence.
Collapse
Affiliation(s)
- Sunny Shinchen Lee
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - Thu Thuy Vũ
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Viet Nam
| | - Anthony S Weiss
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia; Sydney Nano Institute, The University of Sydney, NSW 2006, Australia
| | - Giselle C Yeo
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia.
| |
Collapse
|
57
|
Bellavite P, Fazio S, Affuso F. A Descriptive Review of the Action Mechanisms of Berberine, Quercetin and Silymarin on Insulin Resistance/Hyperinsulinemia and Cardiovascular Prevention. Molecules 2023; 28:4491. [PMID: 37298967 PMCID: PMC10254920 DOI: 10.3390/molecules28114491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Insulin resistance (IR) and the associated hyperinsulinemia are early pathophysiological changes which, if not well treated, can lead to type 2 diabetes, endothelial dysfunction and cardiovascular disease. While diabetes care is fairly well standardized, the prevention and treatment of IR lacks a single pharmaceutical approach and many lifestyle and dietary interventions have been proposed, including a wide range of food supplements. Among the most interesting and well-known natural remedies, alkaloid berberine and the flavonol quercetin have particular relevance in the literature, while silymarin-the active principle of the Silybum marianum thistle-was traditionally used for lipid metabolism disorders and to sustain liver function. This review describes the major defects of insulin signaling leading to IR and the main properties of the three mentioned natural substances, their molecular targets and synergistic action mechanisms. The actions of berberine, quercetin and silymarin are partially superimposable as remedies against reactive oxygen intermediates generated by a high-lipid diet and by NADPH oxidase, which is triggered by phagocyte activation. Furthermore, these compounds inhibit the secretion of a battery of pro-inflammatory cytokines, modulate intestinal microbiota and are especially able to control the various disorders of the insulin receptor and post-receptor signaling systems. Although most of the evidence on the effects of berberine, quercetin and silymarin in modulating insulin resistance and preventing cardiovascular disease derive from experimental studies on animals, the amount of pre-clinical knowledge strongly suggests the need to investigate the therapeutic potential of these substances in human pathology.
Collapse
Affiliation(s)
- Paolo Bellavite
- Pathophysiology Chair, Homeopathic Medical School of Verona, 37121 Verona, Italy
| | - Serafino Fazio
- Department of Internal Medicine, University of Naples Federico II, 80138 Naples, Italy;
| | | |
Collapse
|
58
|
Ding R, Ren X, Sun Q, Sun Z, Duan J. An integral perspective of canonical cigarette and e-cigarette-related cardiovascular toxicity based on the adverse outcome pathway framework. J Adv Res 2023; 48:227-257. [PMID: 35998874 PMCID: PMC10248804 DOI: 10.1016/j.jare.2022.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/29/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Nowadays, cigarette smoking remains the leading cause of chronic disease and premature death, especially cardiovascular disease. As an emerging tobacco product, e-cigarettes have been advocated as alternatives to canonical cigarettes, and thus may be an aid to promote smoking cessation. However, recent studies indicated that e-cigarettes should not be completely harmless to the cardiovascular system. AIM OF REVIEW This review aimed to build up an integral perspective of cigarettes and e-cigarettes-related cardiovascular toxicity. KEY SCIENTIFIC CONCEPTS OF REVIEW This review adopted the adverse outcome pathway (AOP) framework as a pivotal tool and aimed to elucidate the association between the molecular initiating events (MIEs) induced by cigarette and e-cigarette exposure to the cardiovascular adverse outcome. Since the excessive generation of reactive oxygen species (ROS) has been widely approved to play a critical role in cigarette smoke-related CVD and may also be involved in e-cigarette-induced toxic effects, the ROS overproduction and subsequent oxidative stress are regarded as essential parts of this framework. As far as we know, this should be the first AOP framework focusing on cigarette and e-cigarette-related cardiovascular toxicity, and we hope our work to be a guide in exploring the biomarkers and novel therapies for cardiovascular injury.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Xiaoke Ren
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Qinglin Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
59
|
Wang C, Zhao R, Wang Z, Xu T, Huang P. Synthetic ditempolphosphatidylcholine liposome-like nanoparticles for anti-oxidative therapy of atherosclerosis. RSC Adv 2023; 13:16211-16221. [PMID: 37266511 PMCID: PMC10230271 DOI: 10.1039/d3ra01822a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Atherosclerosis (AS), a chronic inflammatory disease, is the leading cause of death worldwide. Anti-oxidative therapy has been developed for AS therapy in light of the critical role of ROS in pathogenesis of AS, but current anti-oxidants have exhibited limited outcomes in the clinic. Herein, new ROS-eliminating liposome-like NPs (Tempol-Lips) were assembled from synthetic lipids that covalently conjugated two Tempol molecules with phosphatidylcholine by esterification reaction. The obtained Tempol-Lips can be efficiently internalized into inflammatory macrophages and attenuated inflammation via scavenging overproduced intracellular ROS. After i.v. administration, Tempol-Lips with nanoscale character accumulated in the plaques of ApoE-/- mice through passive targeting and significantly inhibited the pathogenesis of AS, compared with those treated with control drugs. The therapeutic benefits of Tempol-Lips primarily are ascribed to the reduced local and systematic oxidative stress and inflammation. Preliminary studies in vivo further demonstrated Tempol-Lips were safe and biocompatible after long-term i.v. injection. Conclusively, Tempol-Lips can be developed as a novel anti-AS nanotherapy with potential translation in the clinic.
Collapse
Affiliation(s)
- Chunxiao Wang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| | - Ruifu Zhao
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| | - Zhen Wang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| | - Tingting Xu
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| | - Peng Huang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University Yantai 264000 China
| |
Collapse
|
60
|
Murphy E, Liu JC. Mitochondrial calcium and reactive oxygen species in cardiovascular disease. Cardiovasc Res 2023; 119:1105-1116. [PMID: 35986915 PMCID: PMC10411964 DOI: 10.1093/cvr/cvac134] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 08/11/2023] Open
Abstract
Cardiomyocytes are one of the most mitochondria-rich cell types in the body, with ∼30-40% of the cell volume being composed of mitochondria. Mitochondria are well established as the primary site of adenosine triphosphate (ATP) generation in a beating cardiomyocyte, generating up to 90% of its ATP. Mitochondria have many functions in the cell, which could contribute to susceptibility to and development of cardiovascular disease (CVD). Mitochondria are key players in cell metabolism, ATP production, reactive oxygen species (ROS) production, and cell death. Mitochondrial calcium (Ca2+) plays a critical role in many of these pathways, and thus the dynamics of mitochondrial Ca2+ are important in regulating mitochondrial processes. Alterations in these varied and in many cases interrelated functions play an important role in CVD. This review will focus on the interrelationship of mitochondrial energetics, Ca2+, and ROS and their roles in CVD. Recent insights into the regulation and dysregulation of these pathways have led to some novel therapeutic approaches.
Collapse
Affiliation(s)
- Elizabeth Murphy
- NHLBI, NIH, Bethesda, MD and Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Julia C Liu
- NHLBI, NIH, Bethesda, MD and Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| |
Collapse
|
61
|
Goldoni R, Thomaz DV, Strambini L, Tumedei M, Dongiovanni P, Isola G, Tartaglia G. Quality-by-Design R&D of a Novel Nanozyme-Based Sensor for Saliva Antioxidant Capacity Evaluation. Antioxidants (Basel) 2023; 12:1120. [PMID: 37237985 PMCID: PMC10215665 DOI: 10.3390/antiox12051120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Oxidative stress is one of the main causes of cell damage, leading to the onset of several diseases, and antioxidants represent a barrier against the production of reactive species. Saliva is receiving increasing interest as a promising biofluid to study the onset of diseases and assess the overall health status of an individual. The antioxidant capacity of saliva can be a useful indicator of the health status of the oral cavity, and it is nowadays evaluated mainly through spectroscopic methods that rely on benchtop machines and liquid reagents. We developed a low-cost screen-printed sensor based on cerium oxide nanoparticles that can be used to assess the antioxidant capacity of biofluids as an alternative to traditional methods. The sensor development process was investigated via a quality-by-design approach to identify the most critical parameters of the process for further optimization. The sensor was tested in the detection of ascorbic acid, which is used as an equivalent in the assessment of overall antioxidant capacity. The LoDs ranged from 0.1147 to 0.3528 mM, while the recoveries varied from 80% to 121.1%, being therefore comparable with those of the golden standard SAT test, whose recovery value was 96.3%. Therefore, the sensor achieved a satisfactory sensitivity and linearity in the range of clinical interest for saliva and was validated against the state-of-the-art equipment for antioxidant capacity evaluation.
Collapse
Affiliation(s)
- Riccardo Goldoni
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico Di Milano, 20133 Milan, Italy
- CNR-Istituto di Elettronica e di Ingegneria dell’Informazione e delle Telecomunicazioni, 56122 Pisa, Italy;
| | - Douglas Vieira Thomaz
- National Enterprise for NanoScience and NanoTechnology (NEST), Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
| | - Lucanos Strambini
- CNR-Istituto di Elettronica e di Ingegneria dell’Informazione e delle Telecomunicazioni, 56122 Pisa, Italy;
| | - Margherita Tumedei
- Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, University of Milan, 20100 Milan, Italy;
- UOC Maxillo-Facial Surgery and Dentistry Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy;
| | - Gianluca Tartaglia
- Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, University of Milan, 20100 Milan, Italy;
- UOC Maxillo-Facial Surgery and Dentistry Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
62
|
Faiza N, Imran A, Arshad MU, Arshad MS, Shah MA. Valorization and characterization of corn by-product polyphenols through green extraction technologies. Front Nutr 2023; 10:1107067. [PMID: 37229473 PMCID: PMC10203244 DOI: 10.3389/fnut.2023.1107067] [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: 11/24/2022] [Accepted: 03/27/2023] [Indexed: 05/27/2023] Open
Abstract
The amount of food waste throughout the world has become quite alarming and is contributing to lower food resources. The study aimed to extract and characterize the polyphenols from corn silks at immature and mature stages through conventional and green extraction techniques. Purposely, corn silks, which are some of the by-products of corn, (Zea mays L.) were collected and subjected to proximate analysis including moisture, ash, protein, fiber, and minerals. Secondly, the antioxidants from both immature and mature corn silks were extracted by techniques involving supercritical and ultrasound extraction alongside conventional extraction. The results displayed a promising quantity of protein and fiber along with calcium, magnesium, sodium potassium, and copper. Among the extraction techniques, supercritical extraction at 3,000 Pa acquired the highest total phenolic contents (TPC), total flavonoids (TF), 2, 2-diphenylpicrylhydrazyl (DPPH), ferric-reducing antioxidant power (FRAP) activities as 128.08 ± 3.74 mg GAE/100 g, 86.73 ± 2.75 mg CE/100 g, 106.73 ± 5.10%, and 73.52 ± 2.33 μM Fe + 2/g, respectively, followed by the ultrasound and conventional extraction techniques. Between the immature and mature corn silks, the highest antioxidant activity was displayed by immature corn silks.
Collapse
Affiliation(s)
- Neelam Faiza
- Department of Food Sciences, Government College University, Faisalabad, Pakistan
| | - Ali Imran
- Department of Food Sciences, Government College University, Faisalabad, Pakistan
| | | | | | - Mohd Asif Shah
- Department of Economics, Kebri Dehar University, Kebri Dehar, Ethiopia
- Division of Research and Development, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
63
|
Minjares M, Wu W, Wang JM. Oxidative Stress and MicroRNAs in Endothelial Cells under Metabolic Disorders. Cells 2023; 12:1341. [PMID: 37174741 PMCID: PMC10177439 DOI: 10.3390/cells12091341] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/28/2023] [Accepted: 05/06/2023] [Indexed: 05/15/2023] Open
Abstract
Reactive oxygen species (ROS) are radical oxygen intermediates that serve as important second messengers in signal transduction. However, when the accumulation of these molecules exceeds the buffering capacity of antioxidant enzymes, oxidative stress and endothelial cell (EC) dysfunction occur. EC dysfunction shifts the vascular system into a pro-coagulative, proinflammatory state, thereby increasing the risk of developing cardiovascular (CV) diseases and metabolic disorders. Studies have turned to the investigation of microRNA treatment for CV risk factors, as these post-transcription regulators are known to co-regulate ROS. In this review, we will discuss ROS pathways and generation, normal endothelial cell physiology and ROS-induced dysfunction, and the current knowledge of common metabolic disorders and their connection to oxidative stress. Therapeutic strategies based on microRNAs in response to oxidative stress and microRNA's regulatory roles in controlling ROS will also be explored. It is important to gain an in-depth comprehension of the mechanisms generating ROS and how manipulating these enzymatic byproducts can protect endothelial cell function from oxidative stress and prevent the development of vascular disorders.
Collapse
Affiliation(s)
- Morgan Minjares
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA;
| | - Wendy Wu
- Vera P Shiffman Medical Library, Wayne State University, 320 E Canfield St., Detroit, MI 48201, USA;
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA;
- Center for Molecular Medicine and Genetics, Wayne State University, 320 E Canfield St., Detroit, MI 48201, USA
- Barbara Ann Karmanos Cancer Institute, 4100 John R St., Detroit, MI 48201, USA
| |
Collapse
|
64
|
Zeng X, Xue CD, Li YJ, Qin KR. A mathematical model for intracellular NO and ROS dynamics in vascular endothelial cells activated by exercise-induced wall shear stress. Math Biosci 2023; 359:109009. [PMID: 37086782 DOI: 10.1016/j.mbs.2023.109009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/17/2023] [Accepted: 04/06/2023] [Indexed: 04/24/2023]
Abstract
Vascular endothelial cells (ECs) residing in the innermost layer of blood vessels are exposed to dynamic wall shear stress (WSS) induced by blood flow. The intracellular nitric oxide (NO) and reactive oxygen species (ROS) in ECs modulated by the dynamic WSS play important roles in endothelial functions. Mathematical modeling is a popular methodology for biophysical studies. It can not only explain existing cell experiments, but also reveal the underlying mechanism. However, the previous mathematical models of NO dynamics in ECs are limited to the static WSS induced by constant flow, while arterial blood flow is a periodic pulsatile flow with varying amplitude and frequency at different exercise intensities. In this study, a mathematical model of intracellular NO and ROS dynamics activated by dynamic WSS based on the in vitro cell experiments is developed. With the hypothesis of the viscoelastic body, the Kelvin model is adopted to simulate the mechanosensors on EC. Thus, the NO dynamics activated by dynamic shear stresses induced by constant flow, pulsatile flow, and oscillatory flow are analyzed and compared. Moreover, the roles of ROS have been considered for the first time in the modeling of NO dynamics in ECs based on the analysis of cell experiments. The predictions of the proposed model coincide fairly well with the experimental data when ECs are subjected to exercise-induced WSS. The mechanism is elucidated that WSS induced by moderate-intensity exercise is most favorable to NO production in ECs. This study can provide valuable insights for further study of NO and ROS dynamics in ECs and help develop appropriate exercise regimens for improving endothelial functions.
Collapse
Affiliation(s)
- Xiao Zeng
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian, 116024, Liaoning, PR China.
| | - Chun-Dong Xue
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian, 116024, Liaoning, PR China.
| | - Yong-Jiang Li
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian, 116024, Liaoning, PR China.
| | - Kai-Rong Qin
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, 116024, Liaoning, PR China.
| |
Collapse
|
65
|
Ioannidou S, Kazeli K, Ventouris H, Amanatidou D, Gkinoudis A, Lymperaki E. Correlation of Vitamin 25(OH)D, Liver Enzymes, Potassium, and Oxidative Stress Markers with Lipid Profile and Atheromatic Index: A Pilot Study. J Xenobiot 2023; 13:193-204. [PMID: 37092503 PMCID: PMC10123670 DOI: 10.3390/jox13020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 04/05/2023] Open
Abstract
According to recent literature, there is a limited amount of data about the correlation of vitamin 25(OH)D, potassium (K), oxidative stress parameters, and other biomarkers with dyslipidemia, which is an established risk factor for cardiovascular diseases (CVDs). This study aims to investigate the correlation of lipid profile and atheromatic index TC/HDL with several biomarkers and oxidative stress parameters. A total of 102 volunteers, 67 with atheromatic index TC/HDL > 3.5 (Group A) and 35 with TC/HDL < 3.5 (Group B), aged from 26 to 78 years, participated in this study. Serum levels of triglycerides (TG), total cholesterol (TC), low- and high-density lipoproteins (LDL and HDL), vitamin 25(OH)D [25(OH)D], potassium (K), sodium (Na), lactose dehydrogenase (LDH), liver enzymes including serum glutamic oxaloacetic and glutamic pyruvic transaminases (SGOT and SGPT), gamma-glutamyl transferase (γ-GT), and alkaline phosphatase (ALP) were analyzed using standard photometric methods. Oxidative stress parameters such as reactive oxygen species (ROS) were detected with fluorometric methods, whereas total oxidative (TOS) and antioxidative status (TAS) were measured with spectrophotometric methods. According to the results, negative correlations of HDL (r = −0.593) and 25(OH)D (r = −0.340) and K (r = −0.220) were found, and positive expected correlations of LDL (r = 0.731), TC (r = 0.663), and TG (r = 0.584) with atheromatic index in the total studied sample were found. In conclusion, patients with a dyslipidemic profile should frequently check not only their lipid profile but also other biomarkers such as 25(OH)D, potassium, and oxidative stress markers to predict dyslipidemia and avoid subsequent disorders.
Collapse
Affiliation(s)
- Stavroula Ioannidou
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece
| | - Konstantina Kazeli
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece
- Department of Condensed Matter and Materials Physics, School of Physics, Faculty of Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Hristos Ventouris
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece
| | - Dionysia Amanatidou
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece
| | - Argyrios Gkinoudis
- School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Evgenia Lymperaki
- Department of Biomedical Sciences, International Hellenic University, 57400 Thessaloniki, Greece
| |
Collapse
|
66
|
Jensen KH, Michel BW. Detection of Ethylene with Defined Metal Complexes: Strategies and Recent Advances. ANALYSIS & SENSING 2023; 3:e202200058. [PMID: 37601898 PMCID: PMC10438914 DOI: 10.1002/anse.202200058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Indexed: 08/22/2023]
Abstract
Despite its relative simplicity, ethylene is an interesting molecule with wide-ranging impact in modern chemistry and biology. Stemming from ethylene's role as a critical plant hormone, there has been significant effort to develop selective and sensitive molecular sensors for ethylene. Late transition metal complexes have played an important role in detection strategies due to ethylene's lack of structural complexity and limited reactivity. Two main approaches to ethylene detection are identified: (1) coordination-based sensors, wherein ethylene binds reversibly to a metal center, and (2) activity-based sensors, wherein ethylene undergoes a reaction at a metal center, resulting in the formation and destruction of covalent bonds. Herein, we describe the advantages and disadvantages of various approaches, and the challenges remaining for sensor development.
Collapse
Affiliation(s)
- Katrina H Jensen
- School of Natural Sciences, Black Hills State University, 1200 University Street, Spearfish, SD, 57799, United States
| | - Brian W Michel
- Department of Chemistry and Biochemistry, University of Denver, 2101 E. Wesley Ave, Denver, CO, 80210, United States
| |
Collapse
|
67
|
Wang S, Zhao Y, Chan AWH, Yao M, Chen Z, Abbatt JPD. Organic Peroxides in Aerosol: Key Reactive Intermediates for Multiphase Processes in the Atmosphere. Chem Rev 2023; 123:1635-1679. [PMID: 36630720 DOI: 10.1021/acs.chemrev.2c00430] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Organic peroxides (POs) are organic molecules with one or more peroxide (-O-O-) functional groups. POs are commonly regarded as chemically labile termination products from gas-phase radical chemistry and therefore serve as temporary reservoirs for oxidative radicals (HOx and ROx) in the atmosphere. Owing to their ubiquity, active gas-particle partitioning behavior, and reactivity, POs are key reactive intermediates in atmospheric multiphase processes determining the life cycle (formation, growth, and aging), climate, and health impacts of aerosol. However, there remain substantial gaps in the origin, molecular diversity, and fate of POs due to their complex nature and dynamic behavior. Here, we summarize the current understanding on atmospheric POs, with a focus on their identification and quantification, state-of-the-art analytical developments, molecular-level formation mechanisms, multiphase chemical transformation pathways, as well as environmental and health impacts. We find that interactions with SO2 and transition metal ions are generally the fast PO transformation pathways in atmospheric liquid water, with lifetimes estimated to be minutes to hours, while hydrolysis is particularly important for α-substituted hydroperoxides. Meanwhile, photolysis and thermolysis are likely minor sinks for POs. These multiphase PO transformation pathways are distinctly different from their gas-phase fates, such as photolysis and reaction with OH radicals, which highlights the need to understand the multiphase partitioning of POs. By summarizing the current advances and remaining challenges for the investigation of POs, we propose future research priorities regarding their origin, fate, and impacts in the atmosphere.
Collapse
Affiliation(s)
- Shunyao Wang
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai200444, China
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, OntarioM5S 3E5, Canada
| | - Yue Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
| | - Arthur W H Chan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, OntarioM5S 3E5, Canada
- School of the Environment, University of Toronto, Toronto, OntarioM5S 3E8, Canada
| | - Min Yao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai200240, China
| | - Zhongming Chen
- State Key Laboratory of Environmental Simulation and Pollution Control, College of Environmental Sciences and Engineering, Peking University, Beijing100871, China
| | - Jonathan P D Abbatt
- Department of Chemistry, University of Toronto, Toronto, OntarioM5S 3H6, Canada
| |
Collapse
|
68
|
Wang KX, Ye C, Yang X, Ma P, Yan C, Luo L. New Insights into the Understanding of Mechanisms of Radiation-Induced Heart Disease. Curr Treat Options Oncol 2023; 24:12-29. [PMID: 36598620 DOI: 10.1007/s11864-022-01041-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 01/05/2023]
Abstract
OPINION STATEMENT Cancer patients who receive high-dose thoracic radiotherapy may develop radiation-induced heart disease (RIHD). The clinical presentation of RIHD comprises coronary artery atherosclerosis, valvular disease, pericarditis, cardiomyopathy, and conduction defects. These complications have significantly reduced due to the improved radiotherapy techniques. However, such methods still could not avoid heart radiation exposure. Furthermore, people who received relatively low-dose radiation exposures have exhibited significantly elevated RIHD risks in cohort studies of atomic bomb survivors and occupational exposures. The increased potential in exposure to natural and artificial ionizing radiation sources has emphasized the necessity to understand the development of RIHD. The pathological processes of RIHD include endothelial dysfunction, inflammation, fibrosis, and hypertrophy. The underlying mechanisms may involve the changes in oxidative stress, DNA damage response, telomere erosion, mitochondrial dysfunction, epigenetic regulation, circulation factors, protein post-translational modification, and metabolites. This review will discuss the recent advances in the mechanisms of RIHD at cellular and molecular levels.
Collapse
Affiliation(s)
- Kai-Xuan Wang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Cong Ye
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Xu Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Ping Ma
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, 221004, People's Republic of China
| | - Chen Yan
- Department of Rheumatology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang City, Jiangxi Province, 330006, People's Republic of China.
| | - Lan Luo
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou City, Jiangsu Province, 221004, People's Republic of China.
| |
Collapse
|
69
|
Riaz T, Iqbal MW, Mahmood S, Yasmin I, Leghari AA, Rehman A, Mushtaq A, Ali K, Azam M, Bilal M. Cottonseed oil: A review of extraction techniques, physicochemical, functional, and nutritional properties. Crit Rev Food Sci Nutr 2023; 63:1219-1237. [PMID: 34387525 DOI: 10.1080/10408398.2021.1963206] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Seed oils are the richest source of vitamin-E-active compounds, which contribute significantly to antioxidant activities. Cottonseed oil (CS-O) is attaining more consideration owing to its high fiber content and stability against auto-oxidation. CS-O has gained a good reputation in the global edible oil market due to its distinctive fatty acid profile, anti-inflammatory, and cardio-protective properties. CS-O can be extracted from cottonseed (CS) by microwave-assisted extraction (MAE), aqueous/solvent extraction (A/SE), aqueous ethanol extraction (A-EE), subcritical water extraction, supercritical carbon dioxide extraction (SC-CO2), and enzyme-assisted extraction (E-AE). In this review, the importance, byproducts, physicochemical characteristics, and nutritional profile of CS-O have been explained in detail. This paper also provides a summary of scientific studies existing on functional and phytochemical characteristics of CS-O. Its consumption and health benefits are also deliberated to discover its profitability and applications. CS-O contains 26-35% saturated, 42-52% polyunsaturated, and 18-24% monounsaturated FA. There is approximately 1000 ppm of tocopherols in unprocessed CS-O, but up to one-third is lost during processing. Moreover, besides being consumed as cooking oil, CS-O discovers applications in many fields such as biofuel, livestock, cosmetics, agriculture, and chemicals. This paper provides a comprehensive review of CS-O, its positive benefits, fatty acid profile, extraction techniques, and health applications.HighlightsCS-O is a rich source of exceptional fatty acids.Various techniques to extract the CS-O are discussed.Numerous physicochemical properties of CS-O for the potential market are assessed.It has a wide range of functional properties.Nutritional quality and health benefits are also evaluated.
Collapse
Affiliation(s)
- Tahreem Riaz
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Muhammad Waheed Iqbal
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
- Riphah College of Rehabilitation and Allied Health Sciences, Riphah International University Faisalabad
| | - Shahid Mahmood
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Iqra Yasmin
- Center of Excellence for Olive Research & Training (CEFORT), Barani Agricultural Research Institute (BARI), Chakwal
| | - Ali Ahmad Leghari
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Abdur Rehman
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Anam Mushtaq
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Khubaib Ali
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Muhammad Azam
- Riphah College of Rehabilitation and Allied Health Sciences, Riphah International University Faisalabad
| | - Muhammad Bilal
- Jiangsu Provincial Engineering Laboratory for Biomass Conversion and Process Integration, School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, China
| |
Collapse
|
70
|
Mechanism of inactivation of glyceraldehyde-3-phosphate dehydrogenase in the presence of methylglyoxal. Arch Biochem Biophys 2023; 733:109485. [PMID: 36481268 DOI: 10.1016/j.abb.2022.109485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is known to be one of the targets of methylglyoxal (MGO), a metabolite of glycolysis that increased in diabetes. However, the mechanism of GAPDH inactivation in the presence of MGO is unclear. The purpose of the work was to study the reaction of GAPDH with MGO and to identify the products of the reaction. It was shown that incubation of recombinant human GAPDH with MGO leads to irreversible inactivation of the enzyme, which is accompanied by a decrease in SH-group content by approximately 3.3 per tetramer GAPDH. MALDI-TOF MS analysis showed that the modification of GAPDH with MGO results in the oxidation of the catalytic cysteine residues (Cys152) to form cysteine-sulfinic acid. In addition, 2 arginine residues (R80 and R234) were identified that react with MGO to form hydroimidazolones. Incubation of SH-SY5Y neuroblastoma cells with MGO resulted in the inactivation of GAPDH and inhibition of glycolysis. The mechanism of GAPDH oxidation in the presence of MGO suggests the participation of superoxide anion, which is formed during the reaction of amino groups with methylglyoxal. The role of GAPDH in protection against the damaging effect of ROS in cells in the case of inefficiency of MGO removal by the GSH-dependent glyoxalase system is discussed.
Collapse
|
71
|
Bagheri S, Samiee S, Zarif MN, Deyhim MR. L-carnitine modulates free mitochondrial DNA DAMPs and platelet storage lesions during storage of platelet concentrates. J Thromb Thrombolysis 2023; 55:60-66. [PMID: 36380102 DOI: 10.1007/s11239-022-02725-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2022] [Indexed: 11/16/2022]
Abstract
Platelet storage lesions may occur in Platelet concentrates (PCs) storage time, reducing PCs' quality. Mitochondrial damage causes mitochondrial DNA (mtDNA) to be released into the extracellular space. In this study, we evaluated the effect of L-carnitine (LC) as an antioxidant on free mtDNA DAMPs release in PCs during storage. Ten PCs prepared by the PRP method were studied. The copy numbers of free mtDNA, total reactive oxygen species (ROS), lactate dehydrogenase (LDH) enzyme activity, pH, and platelet counts were measured on days 0, 3, 5, and 7 of PCs storage in LC-treated and untreated platelets. LDH activity was significantly lower than the control group during 7 days of PCs storage (p = 0.041). Also, ROS production decreased in LC-treated PCs compared to the control group during storage (p = 0.026), and the difference mean of ROS between the two groups was significant on day 3, 5, and 7 (Pday3 = 0.02, Pday5 = 0.0001, Pday7 = 0.031). Moreover, LC decreased the copy numbers of free mtDNA during 7 days of storage (p = 0.021), and the difference mean of the copy numbers of free mtDNA in LC-treated PCs compared to the control group was significant on day 5 and 7 (Pday5 = 0.041، Pday7 = 0.022). It seems that LC can maintain the metabolism and antioxidant capacity of PCs and thus can reduce mitochondrial damage and mtDNA release; consequently, it can decrease DAMPs in PCs. Therefore, it may be possible to use this substance as a platelet additive solution in the future.
Collapse
Affiliation(s)
- Saeede Bagheri
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Shahram Samiee
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mahin Nikougoftar Zarif
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mohammad Reza Deyhim
- Department of Clinical Chemistry, Iranian Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, P.O. Box: 14665-1157, Tehran, Iran.
| |
Collapse
|
72
|
Liang Z, Chen Y, Gu R, Guo Q, Nie X. Asiaticoside Prevents Oxidative Stress and Apoptosis in Endothelial Cells by Activating ROS-dependent p53/Bcl-2/Caspase-3 Signaling Pathway. Curr Mol Med 2023; 23:1116-1129. [PMID: 36284389 DOI: 10.2174/1566524023666221024120825] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Asiaticoside (AC) is a triterpenoid saponin found in Centella asiatica (L.) urban extract that has a wide range of pharmacological properties. Our previous study demonstrated that AC could promote angiogenesis in diabetic wounds, but the specific mechanisms remain unknown. OBJECTIVE This study aimed to examine the effectiveness and mechanism of AC on human umbilical vein endothelial cells (HUVECs) exposed to tert-butyl hydroperoxide (t-BHP) toxicity. METHODS Senescence was confirmed using senescence-associated betagalactosidase (SA-β-gal) activity and expression of the cell cycle phase markers p16 and p21. The levels of SOD, NO, MDA, GSH-Px, and ROS were tested. Furthermore, several cell death-related genes and proteins (p53, Bax, Bcl-2 and Caspase-3) were assessed with RT-qPCR and Western blotting. RESULTS AC significantly reduced SA-β-gal activity, with both the suppression of cellcycle inhibitors p16 and p21. We also found that the induced oxidative stress and apoptosis caused by t-BHP treatment resulted in the decrease of antioxidant enzymes activities, the surge of ROS and MDA, the up-regulation of p53, Bax and caspase-3, and the decrease of SOD, NO, GSH-Px and Bcl-2. These biochemical changes were all reversed by treatment with varying doses of AC. CONCLUSION AC alleviates t-BHP-induced oxidative injury and apoptosis in HUVECs through the ROS-dependent p53/Bcl-2/Caspase-3 signaling pathway. It may be a potential antioxidant applied in metabolic disorders and pharmaceutical products.
Collapse
Affiliation(s)
- Zhenwen Liang
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- Key Laboratory of Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563003, China
- College of Pharmacy, Zunyi Medical University, Zunyi 563003, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China
| | - Yu Chen
- Key Laboratory of Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563003, China
- College of Pharmacy, Zunyi Medical University, Zunyi 563003, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China
| | - Rifang Gu
- College of Pharmacy, Zunyi Medical University, Zunyi 563003, China
| | - Qi Guo
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Xuqiang Nie
- Key Laboratory of Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563003, China
- College of Pharmacy, Zunyi Medical University, Zunyi 563003, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China
| |
Collapse
|
73
|
Morrison GC, Eftekhari A, Lakey PSJ, Shiraiwa M, Cummings BE, Waring MS, Williams B. Partitioning of reactive oxygen species from indoor surfaces to indoor aerosols. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2022; 24:2310-2323. [PMID: 36314460 DOI: 10.1039/d2em00307d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Reactive oxygen species (ROS) are among the species thought to be responsible for the adverse health effects of particulate matter (PM) inhalation. Field studies suggest that indoor sources of ROS contribute to measured ROS on PM in indoor air. We hypothesize that ozone reacts on indoor surfaces to form semi-volatile ROS, in particular organic peroxides (OPX), which partition to airborne particles. To test this hypothesis, we modeled ozone-induced formation of OPX, its decay and its partitioning to PM in a residential building and compared the results to field measurements. Simulations indicate that, while ROS of outdoor origin is the primary contributor to indoor ROS (in PM), a substantial fraction of ROS present in indoor PM is from ozone-surface chemistry. At an air change rate equal to 1/h, and an outdoor ozone mixing ratio of 35 ppb, 25% of the ROS concentration in air is due to indoor formation and partitioning of OPX to PM. For the same conditions, but with a modest indoor source of PM (1.5 mg h-1), 44% of indoor ROS on PM is of indoor origin. An indoor source of ozone, such as an electrostatic air cleaner, also increases OPX present in indoor PM. The results of the simulations support the hypothesis that ozone-induced formation of OPX on indoor surfaces, and subsequent partitioning to aerosols, is sufficient to explain field observations. Therefore, indoor sourced ROS could contribute meaningfully to total inhaled PM-ROS.
Collapse
Affiliation(s)
- Glenn C Morrison
- Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, NC, USA.
| | - Azin Eftekhari
- Department of Chemical and Biomolecular Engineering, Georgia Institute of Technology, GA, USA
| | | | - Manabu Shiraiwa
- Department of Chemistry, University of California Irvine, CA, USA
| | - Bryan E Cummings
- Department of Civil, Architectural and Environmental Engineering, Drexel University, Philadelphia, PA, USA
| | - Michael S Waring
- Department of Civil, Architectural and Environmental Engineering, Drexel University, Philadelphia, PA, USA
| | - Brent Williams
- Department of Energy, Environmental and Chemical Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Center for Aerosol Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
74
|
Sato A, Yumita Y, Kagami K, Ishinoda Y, Kimura T, Osaki A, Toya T, Namba T, Endo S, Ido Y, Nagatomo Y, Satoh Y, Adachi T. Endothelial Extracellular Signal-Regulated Kinase/Thromboxane A2/Prostanoid Receptor Pathway Aggravates Endothelial Dysfunction and Insulin Resistance in a Mouse Model of Metabolic Syndrome. J Am Heart Assoc 2022; 11:e027538. [PMID: 36382966 PMCID: PMC9851435 DOI: 10.1161/jaha.122.027538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Metabolic syndrome is characterized by insulin resistance, which impairs intracellular signaling pathways and endothelial NO bioactivity, leading to cardiovascular complications. Extracellular signal-regulated kinase (ERK) is a major component of insulin signaling cascades that can be activated by many vasoactive peptides, hormones, and cytokines that are elevated in metabolic syndrome. The aim of this study was to clarify the role of endothelial ERK2 in vivo on NO bioactivity and insulin resistance in a mouse model of metabolic syndrome. Methods and Results Control and endothelial-specific ERK2 knockout mice were fed a high-fat/high-sucrose diet (HFHSD) for 24 weeks. Systolic blood pressure, endothelial function, and glucose metabolism were investigated. Systolic blood pressure was lowered with increased NO products and decreased thromboxane A2/prostanoid (TP) products in HFHSD-fed ERK2 knockout mice, and Nω-nitro-l-arginine methyl ester (L-NAME) increased it to the levels observed in HFHSD-fed controls. Acetylcholine-induced relaxation of aortic rings was increased, and aortic superoxide level was lowered in HFHSD-fed ERK2 knockout mice. S18886, an antagonist of the TP receptor, improved endothelial function and decreased superoxide level only in the rings from HFHSD-fed controls. Glucose intolerance and the impaired insulin sensitivity were blunted in HFHSD-fed ERK2 knockout mice without changes in body weight. In vivo, S18886 improved endothelial dysfunction, systolic blood pressure, fasting serum glucose and insulin levels, and suppressed nonalcoholic fatty liver disease scores only in HFHSD-fed controls. Conclusions Endothelial ERK2 increased superoxide level and decreased NO bioactivity, resulting in the deterioration of endothelial function, insulin resistance, and steatohepatitis, which were improved by a TP receptor antagonist, in a mouse model of metabolic syndrome.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Yusuke Yumita
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Kazuki Kagami
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Yuki Ishinoda
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Toyokazu Kimura
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Ayumu Osaki
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Takumi Toya
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Takayuki Namba
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Shogo Endo
- Department of Aging NeuroscienceTokyo Metropolitan Institute of GerontologyTokyoJapan
| | - Yasuo Ido
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Yuji Nagatomo
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| | - Yasushi Satoh
- Department of BiochemistryNational Defense Medical CollegeTokorozawaJapan
| | - Takeshi Adachi
- Department of CardiologyNational Defense Medical CollegeTokorozawaJapan
| |
Collapse
|
75
|
Carbonic Anhydrase inhibitors bearing organotelluride moieties as novel agents for antitumor therapy. Eur J Med Chem 2022; 244:114811. [DOI: 10.1016/j.ejmech.2022.114811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/23/2022]
|
76
|
Jia Z, Song R, Xu Y, Liu X, Zhang X. Astaxanthin absorption modulated antioxidant enzyme activity and targeted specific metabolic pathways in rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:7003-7016. [PMID: 35689476 DOI: 10.1002/jsfa.12062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 01/25/2022] [Accepted: 06/11/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Saponification contributed to an increase in the in vitro antioxidant activity of astaxanthin (Asta) extracts derived from Penaeus sinensis (Solenocera crassicornis) by-products. However, the influence of non-saponification (N-Asta) and saponification Asta (S-Asta) absorption on antioxidant activity in vivo was limited. The antioxidant properties of N-Asta and S-Asta were therefore compared in Sprague Dawley male rats after 6 h and 12 of absorption using biochemistry assays combined with an untargeted metabonomics strategy. RESULTS Non-saponified Asta and S-Asta showed similar digestive properties in a stimulated gastrointestinal tract. Increased glutathione content and decreased malondialdehyde content were measured in the liver tissues of N-Asta and S-Asta treated rats after 12 h of absorption. Absorption of N-Asta increased liver total superoxide dismutase, glutathione peroxidase, and catalase activity. Treatment with S-Asta up-regulated NAD(P)H: quinine oxidoreductase-1, and heme oxygenase-1 expression was associated with the nuclear erythroid 2-related factor 2/antioxidant responsive element pathway at the end of 12 h absorption. With partial least square-discriminant analysis and metabolite heatmap profiles, the S-Asta group was clearly separated from the N-Asta group. The S-Asta treatment also demonstrated stronger influences on plasma metabolites than the N-Asta treatment. Both N-Asta and S-Asta absorption showed critical roles in the regulation of specific metabolites, and 15 potential biomarkers were identified in eight key pathways to separate these experimental groups after 12 h of absorption. However, an increased serotonin level was only detected in the S-Asta group after 12 h absorption. CONCLUSION Absorption of N-Asta and S-Asta induced different antioxidant effects in normal rats, which were associated with metabolite changes. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhe Jia
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, School of Food Science and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Ru Song
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, School of Food Science and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Yan Xu
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, School of Food Science and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Xinyan Liu
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, School of Food Science and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Xiaoxia Zhang
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, School of Food Science and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| |
Collapse
|
77
|
Kumar VHS. Cardiovascular Morbidities in Adults Born Preterm: Getting to the Heart of the Matter! CHILDREN (BASEL, SWITZERLAND) 2022; 9:children9121843. [PMID: 36553286 PMCID: PMC9777245 DOI: 10.3390/children9121843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
Advances in perinatal and neonatal care have led to improved survival of preterm infants into adulthood. However, the shift in focus to long-term health in adults born preterm requires a clear understanding of the impact of prematurity on developing organ systems and the development of adult-oriented disease. A less well-recognized area of risk for surviving preterm infants is their cardiometabolic health. Epidemiologic evidence has linked preterm birth to the development of systemic hypertension, type 2 diabetes, metabolic syndrome, heart failure, and ischemic heart disease. Of more significant concern is that the risk of cardiometabolic disorders is higher in adults born preterm compared to full-term infants. The interconnected nature of the cardio-pulmonary system means worsening morbidity and mortality in adults born preterm. Addressing the problems of adults born preterm holistically would help promote cardiovascular health, wellness, and quality of life over their lifetime. Recognizing that adults born preterm are a unique subset of the population is a challenge in the current healthcare environment. Addressing issues relevant to adults born preterm in the clinically and research domain, using technology to characterize cardiopulmonary physiology and exercise tolerance, developing screening tools for early diagnosis and treatment, and robust follow-up of these infants with access to longitudinal data would improve both the quality and longevity of life in adults born preterm.
Collapse
Affiliation(s)
- Vasantha H S Kumar
- Division of Neonatology, Department of Pediatrics, John R Oishei Children's Hospital, University at Buffalo, 1001 5th Floor Main Street, Buffalo, NY 14203, USA
| |
Collapse
|
78
|
P66Shc (Shc1) Zebrafish Mutant Line as a Platform for Testing Decreased Reactive Oxygen Species in Pathology. J Cardiovasc Dev Dis 2022; 9:jcdd9110385. [DOI: 10.3390/jcdd9110385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
Reactive oxygen species (ROS) dysregulation exacerbates many pathologies but must remain within normal ranges to maintain cell function. Since ROS-mediated pathology and routine cell function are coupled, in vivo models evaluating low-ROS background effects on pathology are limited. Some models alter enzymatic antioxidant expression/activity, while others involve small molecule antioxidant administration. These models cause non-specific ROS neutralization, decreasing both beneficial and detrimental ROS. This is detrimental in cardiovascular pathology, despite the negative effects excessive ROS has on these pathologies. Thus, current trends in ROS-mediated pathology have shifted toward selective inhibition of ROS producers that are dysregulated during pathological insults, such as p66Shc. In this study, we evaluated a zebrafish heterozygote p66Shc hypomorphic mutant line as a low-ROS myocardial infarction (MI) pathology model that mimics mammalian MI. Our findings suggest this zebrafish line does not have an associated negative phenotype, but has decreased body mass and tissue ROS levels that confer protection against ROS-mediated pathology. Therefore, this line may provide a low-ROS background leading to new insights into disease.
Collapse
|
79
|
Xu H, Zhang Z, Zhang L, Chen Z, Wang S. Tungsten disulfide nanoflowers with multi-nanoenzyme activities for the treatment of acute liver injury. J Colloid Interface Sci 2022; 625:544-554. [PMID: 35749849 DOI: 10.1016/j.jcis.2022.06.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/04/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022]
Abstract
In this study, polyvinyl pyrrolidone modified tungsten disulfide (WS2-PVP) nanoflower was synthesized using a simple and effective one-pot method. Owing to the surface polyvinyl pyrrolidone (PVP) modification, WS2-PVP nanoflowers showed excellent colloidal stability in different circumstances, which can be well dispersed in water, saline, and cell culture medium. Meanwhile, the WS2-PVP nanoflowers have a good biocompatibility both in vitro and in vivo. Further studies confirmed that the WS2-PVP nanoflowers have the ability of simulating catalase, superoxide dismutase and glutathione peroxidase enzymes and scavenging reactive oxygen species (ROS). Therefore, WS2-PVP nanoflowers were used to treat reactive oxygen species-related diseases, which showed the cell protection effect and significantly improved the treatment results of acute liver injury on mice. We hope that our findings will facilitate the development of nanomaterials with multiple enzymatic mimicking properties and further clinical application of tungsten-based ROS scavengers in biomedical therapy and research.
Collapse
Affiliation(s)
- Hao Xu
- Department of Infectious Diseases, Changhai Hospital, Naval Military Medical University, Shanghai 200433, China
| | - Zhirui Zhang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, China
| | - Liying Zhang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, China
| | - Zheng Chen
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, China
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, China.
| |
Collapse
|
80
|
Yang L, Guo H, Hou T, Li F. Uncovering the Interaction between Intracellular Telomerase Activity and Hydrogen Peroxide during Cancer Cell Apoptosis Utilizing a Dual-Color Fluorescent Nanoprobe. Anal Chem 2022; 94:15162-15169. [PMID: 36256448 DOI: 10.1021/acs.analchem.2c03695] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Uncovering the intrinsic interaction of different bioactive species, i.e., reactive oxygen species (ROS) and telomerase, is of great importance because they play interrelated and interdependent biological roles in living organisms. Nevertheless, exploration of the intracellular ROS/telomerase cross-talk by effective and noninvasive methods remains a great challenge, as it is difficult to simultaneously detect different types of biomolecules (i.e., active small molecules and proteins) in living cells. To address this issue, herein, we report, for the first time, a novel fluorescent nanoprobe for simultaneous determination and in situ imaging of telomerase activity and hydrogen peroxide (H2O2) in living cells. With the advantage of high sensitivity and good specificity, this newly fabricated nanoprobe was successfully applied to precisely visualize and monitor the changes in telomerase activity and H2O2 concentration in cancer cells. More significantly, by employing the nanoprobe as a one-step incubation tool, it is found that there is a cross-talk between H2O2 and telomerase activity in the drug-induced cancer cells' apoptosis process, which provides valuable information for gaining fundamental insights into the relationship between ROS and telomerase activity in cancer treatments. This work affords a promising method for revealing the relevant regulatory mechanisms and roles of ROS and telomerase activity in the occurrence, evolvement, and treatment of diseases.
Collapse
Affiliation(s)
- Limin Yang
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Heng Guo
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Ting Hou
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| | - Feng Li
- College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao 266109, People's Republic of China
| |
Collapse
|
81
|
Development of HPTLC method for the simultaneous estimation of quercetin, curcumin, and ascorbic acid in herbal formulations. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2022. [PMCID: PMC9148195 DOI: 10.1007/s13738-022-02586-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Antioxidants can scavenge free radicals in the human body and have been associated with a protective effect on the human body against a variety of diseases. As a part of normal cellular function, free radical production occurs continuously within the body at a cellular level. Excess-free radical production from endogenous or exogenous sources may result in a variety of diseases. Vitamins (A, C, and E), carotenoids, polyphenols, and flavonoids are some examples of common natural antioxidants. Among the different antioxidants quercetin, curcumin, and ascorbic acids have been extensively studied, and numerous reports on their antioxidant activity are available in the literature. Antioxidants are regarded as a key player in the current pandemic situation, and they play a key role as a preventive and controlling tool. There are a variety of herbal formulations and immune booster therapies available, many of which claim to contain various antioxidants and are effective against covid-19. Numerous herbal formulations and immune booster therapies are available and claimed to have different antioxidants and other constituents. In the present study simple, rapid, accurate, precise, and sensitive high-performance thin-layer chromatographic analytical method was developed for simultaneous estimation of quercetin, curcumin, and ascorbic acid. The developed method can effectively employ for different polyherbal formulations. The developed method was validated according to ICH guidelines. In the present study, an aluminum plate precoated with 60F 254 silica gel was used as a stationary phase whereas chloroform: ethyl acetate: formic acid (6:6:2.5 v/v/v) was used as a mobile phase. Chromatographic detection was performed with a Camag TLC scanner at 265 nm. The linearity range for the proposed method was found to be 500–1000 ng/band for all three analytes. The antioxidant potentiality of selected antioxidants was evaluated by an optimized UV visible spectroscopic-reducing power method. The reducing power method was optimized for the concentration and volume of reagent, incubation temperature, and time. The results of the study revealed that quercetin is having the best antioxidant potential compared to curcumin and ascorbic acid.
Collapse
|
82
|
Zhao K, Xu T, Mao Y, Wu X, Hua D, Sheng Y, Li P. Alamandine alleviated heart failure and fibrosis in myocardial infarction mice. Biol Direct 2022; 17:25. [PMID: 36167556 PMCID: PMC9516792 DOI: 10.1186/s13062-022-00338-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/20/2022] [Indexed: 11/10/2022] Open
Abstract
Alamandine (Ala) is the newest identified peptide of the renin-angiotensin system and has protective effect on myocyte hypertrophy. However, it is still unclear whether Ala can alleviate heart failure (HF). The aim of this study was to explore the effects of Ala on HF and the related cardiac fibrosis, and to probe the mechanism. HF model was induced by myocardial infarction (MI) in mice. Four weeks after MI, Ala was administrated by intraperitoneal injection for two weeks. Ala injection significantly improved cardiac dysfunction of MI mice in vivo. The cardiac fibrosis and the related biomarkers were attenuated after Ala administration in HF mice in vivo. The increases of collagen I, alpha-smooth muscle actin and transforming growth factor-beta induced by oxygen–glucose deprivation (OGD) in neonatal rat cardiac fibroblasts (NRCFs) were inhibited by Ala treatment in vitro. The biomarkers of apoptosis were elevated in NRCFs induced by OGD, which were attenuated after treating with Ala in vitro. The enhancement of oxidative stress in the heart of MI mice or in the NRCFs treated with OGD was suppressed by treating with Ala in vivo and in vitro. These effects of Ala were reversed by tBHP, an exogenous inducer of oxidative stress in vitro. These results demonstrated that Ala could alleviate cardiac dysfunction and attenuate cardiac fibrosis via inhibition of oxidative stress.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Tianhua Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yukang Mao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Xiaoguang Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Dongxu Hua
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yanhui Sheng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China. .,Department of Cardiology, Jiangsu Province Hospital, Nanjing, Jiangsu, China.
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
83
|
Pandey AK, Waldeck-Weiermair M, Wells QS, Xiao W, Yadav S, Eroglu E, Michel T, Loscalzo J. Expression of CD70 Modulates Nitric Oxide and Redox Status in Endothelial Cells. Arterioscler Thromb Vasc Biol 2022; 42:1169-1185. [PMID: 35924558 PMCID: PMC9394499 DOI: 10.1161/atvbaha.122.317866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endothelial dysfunction is a critical component in the pathogenesis of cardiovascular diseases and is closely associated with nitric oxide (NO) levels and oxidative stress. Here, we report on novel findings linking endothelial expression of CD70 (also known as CD27 ligand) with alterations in NO and reactive oxygen species. METHODS CD70 expression was genetically manipulated in human aortic and pulmonary artery endothelial cells. Intracellular NO and hydrogen peroxide (H2O2) were measured using genetically encoded biosensors, and cellular phenotypes were assessed. RESULTS An unbiased phenome-wide association study demonstrated that polymorphisms in CD70 associate with vascular phenotypes. Endothelial cells treated with CD70-directed short-interfering RNA demonstrated impaired wound closure, decreased agonist-stimulated NO levels, and reduced eNOS (endothelial nitric oxide synthase) protein. These changes were accompanied by reduced NO bioactivity, increased 3-nitrotyrosine levels, and a decrease in the eNOS binding partner heat shock protein 90. Following treatment with the thioredoxin inhibitor auranofin or with agonist histamine, intracellular H2O2 levels increased up to 80% in the cytosol, plasmalemmal caveolae, and mitochondria. There was increased expression of NADPH oxidase 1 complex and gp91phox; expression of copper/zinc and manganese superoxide dismutases was also elevated. CD70 knockdown reduced levels of the H2O2 scavenger catalase; by contrast, glutathione peroxidase 1 expression and activity were increased. CD70 overexpression enhanced endothelial wound closure, increased NO levels, and attenuated the reduction in eNOS mRNA induced by TNFα. CONCLUSIONS Taken together, these data establish CD70 as a novel regulatory protein in endothelial NO and reactive oxygen species homeostasis, with implications for human vascular disease.
Collapse
Affiliation(s)
- Arvind K. Pandey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA (A.K.P., M.W.-W., W.X., S.Y., T.M., J.L.)
| | - Markus Waldeck-Weiermair
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA (A.K.P., M.W.-W., W.X., S.Y., T.M., J.L.)
| | - Quinn S. Wells
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, TN (Q.S.W.)
| | - Wusheng Xiao
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA (A.K.P., M.W.-W., W.X., S.Y., T.M., J.L.)
| | - Shambhu Yadav
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA (A.K.P., M.W.-W., W.X., S.Y., T.M., J.L.)
| | - Emrah Eroglu
- Faculty for Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey (E.E.)
| | - Thomas Michel
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA (A.K.P., M.W.-W., W.X., S.Y., T.M., J.L.)
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA (A.K.P., M.W.-W., W.X., S.Y., T.M., J.L.)
| |
Collapse
|
84
|
Zhang Z, Dalan R, Hu Z, Wang JW, Chew NW, Poh KK, Tan RS, Soong TW, Dai Y, Ye L, Chen X. Reactive Oxygen Species Scavenging Nanomedicine for the Treatment of Ischemic Heart Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202169. [PMID: 35470476 DOI: 10.1002/adma.202202169] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Ischemic heart disease (IHD) is the leading cause of disability and mortality worldwide. Reactive oxygen species (ROS) have been shown to play key roles in the progression of diabetes, hypertension, and hypercholesterolemia, which are independent risk factors that lead to atherosclerosis and the development of IHD. Engineered biomaterial-based nanomedicines are under extensive investigation and exploration, serving as smart and multifunctional nanocarriers for synergistic therapeutic effect. Capitalizing on cell/molecule-targeting drug delivery, nanomedicines present enhanced specificity and safety with favorable pharmacokinetics and pharmacodynamics. Herein, the roles of ROS in both IHD and its risk factors are discussed, highlighting cardiovascular medications that have antioxidant properties, and summarizing the advantages, properties, and recent achievements of nanomedicines that have ROS scavenging capacity for the treatment of diabetes, hypertension, hypercholesterolemia, atherosclerosis, ischemia/reperfusion, and myocardial infarction. Finally, the current challenges of nanomedicines for ROS-scavenging treatment of IHD and possible future directions are discussed from a clinical perspective.
Collapse
Affiliation(s)
- Zhan Zhang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 408433, Singapore
| | - Zhenyu Hu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jiong-Wei Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Nicholas Ws Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 119609, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macao, Taipa, Macau SAR, 999078, China
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Chemical and Biomolecular Engineering and Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
85
|
Xu H, She P, Ma B, Zhao Z, Li G, Wang Y. ROS responsive nanoparticles loaded with lipid-specific AIEgen for atherosclerosis-targeted diagnosis and bifunctional therapy. Biomaterials 2022; 288:121734. [PMID: 35999079 DOI: 10.1016/j.biomaterials.2022.121734] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022]
Abstract
Atherosclerosis, which is triggered by endothelial damage, progressive local inflammation and excessive lipid accumulation, is one of the most common cardiovascular diseases in recent years. Drug delivery systems have shown great potential for the accurate diagnosis and effective treatment of early atherosclerosis, but are accompanied by disadvantages such as poor stability, lack of active targeting and non-specific recognition capabilities, which still need to be further developed. In our work, a multifunctional nanoparticle (LFP/PCDPD) with reactive oxygen species (ROS) responsive drug release, lipid removal, and lipid-specific AIE fluorescence imaging was constructed. Cyclodextrin structure with lipid removal function and PMEMA blocks with ROS-response-mediated hydrophobic to hydrophilic conversion were simultaneously introduced into the structure of LFP/PCDPD to load the anti-inflammatory drug prednisolone (Pred) and lipid-specific AIEgen (LFP). The active targeting function of LFP/PCDPD was conferred by the high affinity of dextran to the vascular adhesion molecule-1 (VCAM-1) and CD44 receptor on the surface of broken endothelial cells. After intravenous injection into ApoE-/- mice, LFP/PCDPD actively enriched in the microenvironment of local ROS overexpression and rich lipids in atherosclerosis. Pred and LFP were released while lipids were removed, thus enabling proactive targeting of atherosclerosis and efficient "two-pronged" treatment.
Collapse
Affiliation(s)
- Hong Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Peiyi She
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Boxuan Ma
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, 310016, China
| | - Zhiyu Zhao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
86
|
CHEN L, GAO W, SHAO Y, LI C, LU Y. Alpha-lipoic acid prevents atrial electrical and structural remodeling via inhibition of NADPH oxidase in a rabbit rapid atrial pacing model. Turk J Med Sci 2022; 52:1378-1388. [PMID: 36326363 PMCID: PMC10387899 DOI: 10.55730/1300-0144.5445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 08/10/2022] [Accepted: 03/19/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Alpha-lipoic acid (ALA) is a natural compound, one of the natural antioxidants with high activity. In the NADPH oxidase family, NADPH oxidase 4 (NOX4) is an important subunit participating in the production of ROS. NADPH oxidase 2 (NOX2) can form active NADPH oxidase complexes when binding to several other subunits in the cytoplasm, and NOX2 is its major functional subunit. Rapid atrial pacing (RAP) model was constructed to study the effects of ALA on electrical and structural remodeling in rabbits. METHODS Thirty rabbits were divided into SHAM group, RAP group and ALA+RAP group. Their right atriums were paced at a speed of 600 beats/min for 12 h in the RAP and ALA+RAP groups, and the atrial effective refractory period (AERP) and AERP frequency adaptability were determined during the pace. In ALA+RAP group, ALA (30 mg/kg) was administered intraperitoneally daily to the rabbits for 3 days before RAP. Atrial tissue was collected from each group to detect malondialdehyde (MDA), superoxide dismutase (SOD) and reactive oxygen species (ROS) to observe the effect of oxidative stress. The pathological structure of the atrial tissue was observed through hematoxylin-eosin (HE) staining. Ultrastructural changes in the atrial myocytes were observed by transmission electron microscopy (TEM), and the expression levels of Nox2 and Nox4 were detected by immunohistochemistry, western blot and ELISA. RESULTS AERP gradually shortened, while ALA injection could remarkably delay this process. HE staining showed that the most of the nuclei appeared normal, the myocardial fibers did not show ruptures, and their arrangement was slightly ordered, and myofilament dissolution and mitochondrial swelling and deformation were rarely observed by TEM in the ALA+RAP group. Compared with the RAP group, the contents of MDA and ROS were reduced, SOD activity was enhanced, and the expression of NOX2 and NOX4 was decreased in the ALA+RAP group. DISCUSSION ALA can inhibit atrial electrical remodeling and structural remodeling, and its mechanism may be related to inhibiting the activity of NADPH oxidase.
Collapse
Affiliation(s)
- Lei CHEN
- Department of Cardiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou,
China
| | - Wen GAO
- Department of Cardiology, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou,
China
| | - Yameng SHAO
- Department of Cardiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou,
China
| | - Chenggang LI
- Department of Cardiology, Xuzhou New Health Geriatric Hospital, Xuzhou,
China
| | - Yuan LU
- Department of Cardiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou,
China
| |
Collapse
|
87
|
Li Z, Guo X, Qin J, Guan Y, Li J, Guo H, Zhang B, Zhang M, Tang J. Size-effect on the intracellular antioxidative activity of Prussian blue nanoparticles investigated by atomic force microscopy. Anal Chim Acta 2022; 1227:340321. [DOI: 10.1016/j.aca.2022.340321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022]
|
88
|
Glycolysis and the Pentose Phosphate Pathway Promote LPS-Induced NOX2 Oxidase- and IFN-β-Dependent Inflammation in Macrophages. Antioxidants (Basel) 2022; 11:antiox11081488. [PMID: 36009206 PMCID: PMC9405479 DOI: 10.3390/antiox11081488] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages undergo a metabolic switch from oxidative phosphorylation to glycolysis when exposed to gram-negative bacterial lipopolysaccharide (LPS), which modulates antibacterial host defence mechanisms. Here, we show that LPS treatment of macrophages increased the classical oxidative burst response via the NADPH oxidase (NOX) 2 enzyme, which was blocked by 2-deoxyglucose (2-DG) inhibition of glycolysis. The inhibition of the pentose phosphate pathway with 6-aminonicotinamide (6-AN) also suppressed the LPS-induced increase in NOX2 activity and was associated with a significant reduction in the mRNA expression of NOX2 and its organizer protein p47phox. Notably, the LPS-dependent enhancement in NOX2 oxidase activity was independent of both succinate and mitochondrial reactive oxygen species (ROS) production. LPS also increased type I IFN-β expression, which was suppressed by 2-DG and 6-AN and, therefore, is dependent on glycolysis and the pentose phosphate pathway. The type I IFN-β response to LPS was also inhibited by apocynin pre-treatment, suggesting that NOX2-derived ROS promotes the TLR4-induced response to LPS. Moreover, recombinant IFN-β increased NOX2 oxidase-dependent ROS production, as well as NOX2 and p47phox expression. Our findings identify a previously undescribed molecular mechanism where both glycolysis and the pentose phosphate pathway are required to promote LPS-induced inflammation in macrophages.
Collapse
|
89
|
Zhang Q, Wang L, Yin Y, Shen J, Xie J, Yuan J. Hydrogen sulfide releasing hydrogel for alleviating cardiac inflammation and protecting against myocardial ischemia-reperfusion injury. J Mater Chem B 2022; 10:5344-5351. [PMID: 35792619 DOI: 10.1039/d2tb00971d] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Myocardial infarction is one of the leading causes of death worldwide. Thus, protection against myocardial ischemia-reperfusion injury is particularly important to improve the prognosis of myocardial infarction. Recently, hydrogen sulfide (H2S) has been reported to possess a protective effect against myocardial ischemia-reperfusion injury. However, an effective gas delivery system to release H2S controllably at an appropriate concentration needs to be further investigated. In this study, a new H2S-releasing hydrogel system was constructed and applied in an experimental I/R model of rats. The administration of the hydrogel significantly ameliorated microvascular obstruction, prevented myocardial fibrosis, and attenuated cardiac inflammation. This suggested that the novel H2S-releasing hydrogel represented a promising therapeutic strategy targeting myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Cardiology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, 210093, China.
| | - Lijuan Wang
- National and Local Joint Engineering Research Center of Biofunctional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.
| | - Yong Yin
- Department of Cardiology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, 210093, China.
| | - Jian Shen
- National and Local Joint Engineering Research Center of Biofunctional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.
| | - Jun Xie
- Department of Cardiology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing, 210093, China.
| | - Jiang Yuan
- National and Local Joint Engineering Research Center of Biofunctional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
90
|
Atherosclerosis Plaque Reduction by Lycopene Is Mediated by Increased Energy Expenditure through AMPK and PPARα in ApoE KO Mice Fed with a High Fat Diet. Biomolecules 2022; 12:biom12070973. [PMID: 35883529 PMCID: PMC9313394 DOI: 10.3390/biom12070973] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 02/04/2023] Open
Abstract
Lycopene is a carotenoid found in tomatoes that has potent antioxidant activity. The Mediterranean diet is particularly rich in lycopene, which has well-known beneficial effects on cardiovascular health. We tested the effects of lycopene extract in a group of 20 ApoE knockout mice, fed with a high fat western diet for 14 weeks. Starting from week 3 and up to week 14, the mice were randomly divided into two groups that received lycopene (n = 10) by oral suspension every day at the human equivalent dose of 60 mg/day (0.246 mg/mouse/day), or the vehicle solution (n = 10). The lycopene administration reduced triglycerides and cholesterol blood levels starting from week 6 and continuing through to the end of the experiment (p < 0.001). This reduction was mediated by an enhanced liver expression of PPAR-α and AMPK-α and reduced SREBP levels (p < 0.0001). As a histological red-out, the extent of atherosclerotic plaques and the intima−media thickness in the aorta were significantly reduced by lycopene. In this context, lycopene augmented the Nrf-2 positivity staining in the endothelium, thereby confirming that its antioxidant activity was mediated by this nuclear factor. The positive results obtained in this pre-clinical model further support the use of lycopene extracts to reduce atherosclerosis.
Collapse
|
91
|
Ma Q, Xu S, Zhai Z, Wang K, Liu X, Xiao H, Zhuo S, Liu Y. Recent Progress of Small‐Molecule Ratiometric Fluorescent Probes for Peroxynitrite in Biological Systems. Chemistry 2022; 28:e202200828. [DOI: 10.1002/chem.202200828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Qingqing Ma
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Shanlin Xu
- Department of Oncology, Zibo Central Hospital Zibo 255036 P. R. China
| | - Zhaodong Zhai
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Kai Wang
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Xueli Liu
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Haibin Xiao
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Shuping Zhuo
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| | - Yuying Liu
- School of Chemistry and Chemical Engineering Shandong University of Technology Zibo 255049 P. R. China
| |
Collapse
|
92
|
Hu Q, Yammani RD, Brown-Harding H, Soto-Pantoja DR, Poole LB, Lukesh JC. Mitigation of doxorubicin-induced cardiotoxicity with an H2O2-Activated, H2S-Donating hybrid prodrug. Redox Biol 2022; 53:102338. [PMID: 35609400 PMCID: PMC9126844 DOI: 10.1016/j.redox.2022.102338] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/02/2022] [Accepted: 05/11/2022] [Indexed: 01/25/2023] Open
Affiliation(s)
- Qiwei Hu
- Department of Chemistry, Wake Forest University, Wake Downtown Campus, Winston-Salem, NC, 27101, USA
| | - Rama D Yammani
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | | | - David R Soto-Pantoja
- Department of Cancer Biology and Department of Surgery/Hypertension, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Leslie B Poole
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| | - John C Lukesh
- Department of Chemistry, Wake Forest University, Wake Downtown Campus, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
93
|
Lansdell TA, Chambers LC, Dorrance AM. Endothelial Cells and the Cerebral Circulation. Compr Physiol 2022; 12:3449-3508. [PMID: 35766836 DOI: 10.1002/cphy.c210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells form the innermost layer of all blood vessels and are the only vascular component that remains throughout all vascular segments. The cerebral vasculature has several unique properties not found in the peripheral circulation; this requires that the cerebral endothelium be considered as a unique entity. Cerebral endothelial cells perform several functions vital for brain health. The cerebral vasculature is responsible for protecting the brain from external threats carried in the blood. The endothelial cells are central to this requirement as they form the basis of the blood-brain barrier. The endothelium also regulates fibrinolysis, thrombosis, platelet activation, vascular permeability, metabolism, catabolism, inflammation, and white cell trafficking. Endothelial cells regulate the changes in vascular structure caused by angiogenesis and artery remodeling. Further, the endothelium contributes to vascular tone, allowing proper perfusion of the brain which has high energy demands and no energy stores. In this article, we discuss the basic anatomy and physiology of the cerebral endothelium. Where appropriate, we discuss the detrimental effects of high blood pressure on the cerebral endothelium and the contribution of cerebrovascular disease endothelial dysfunction and dementia. © 2022 American Physiological Society. Compr Physiol 12:3449-3508, 2022.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
94
|
Panda P, Verma HK, Lakkakula S, Merchant N, Kadir F, Rahman S, Jeffree MS, Lakkakula BVKS, Rao PV. Biomarkers of Oxidative Stress Tethered to Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9154295. [PMID: 35783193 PMCID: PMC9249518 DOI: 10.1155/2022/9154295] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease (CVD) is a broad term that incorporated a group of conditions that affect the blood vessels and the heart. CVD is a foremost cause of fatalities around the world. Multiple pathophysiological mechanisms are involved in CVD; however, oxidative stress plays a vital role in generating reactive oxygen species (ROS). Oxidative stress occurs when the concentration of oxidants exceeds the potency of antioxidants within the body while producing reactive nitrogen species (RNS). ROS generated by oxidative stress disrupts cell signaling, DNA damage, lipids, and proteins, thereby resulting in inflammation and apoptosis. Mitochondria is the primary source of ROS production within cells. Increased ROS production reduces nitric oxide (NO) bioavailability, which elevates vasoconstriction within the arteries and contributes to the development of hypertension. ROS production has also been linked to the development of atherosclerotic plaque. Antioxidants can decrease oxidative stress in the body; however, various therapeutic drugs have been designed to treat oxidative stress damage due to CVD. The present review provides a detailed narrative of the oxidative stress and ROS generation with a primary focus on the oxidative stress biomarker and its association with CVD. We have also discussed the complex relationship between inflammation and endothelial dysfunction in CVD as well as oxidative stress-induced obesity in CVD. Finally, we discussed the role of antioxidants in reducing oxidative stress in CVD.
Collapse
Affiliation(s)
- Poojarani Panda
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lung Biology and Disease, Helmholtz Zentrum, 85764 Neuherberg, Munich, Germany
| | | | - Neha Merchant
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali, 304022 Rajasthan, India
| | - Fairrul Kadir
- Department of Emergency Medicine, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Shamsur Rahman
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Mohammad Saffree Jeffree
- Department of Community and Family Medicine, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, 88400 Sabah, Malaysia
| | | | - Pasupuleti Visweswara Rao
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Abdurrab University, Jalan Riau Ujung No. 73, Pekanbaru, 28292 Riau, Indonesia
- Centre for International Relations and Research Collaborations, Reva University, Rukmini Knowledge Park, Kattigenahalli, Yelahanka, Bangalore, 560064 Karnataka, India
| |
Collapse
|
95
|
Haslem L, Hays JM, Hays FA. p66Shc in Cardiovascular Pathology. Cells 2022; 11:cells11111855. [PMID: 35681549 PMCID: PMC9180016 DOI: 10.3390/cells11111855] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/06/2023] Open
Abstract
p66Shc is a widely expressed protein that governs a variety of cardiovascular pathologies by generating, and exacerbating, pro-apoptotic ROS signals. Here, we review p66Shc’s connections to reactive oxygen species, expression, localization, and discuss p66Shc signaling and mitochondrial functions. Emphasis is placed on recent p66Shc mitochondrial function discoveries including structure/function relationships, ROS identity and regulation, mechanistic insights, and how p66Shc-cyt c interactions can influence p66Shc mitochondrial function. Based on recent findings, a new p66Shc mitochondrial function model is also put forth wherein p66Shc acts as a rheostat that can promote or antagonize apoptosis. A discussion of how the revised p66Shc model fits previous findings in p66Shc-mediated cardiovascular pathology follows.
Collapse
Affiliation(s)
- Landon Haslem
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Jennifer M. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Franklin A. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
- Stephenson Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Correspondence:
| |
Collapse
|
96
|
Discrepancy between the Actions of Glucagon-like Peptide-1 Receptor Ligands in the Protection of the Heart against Ischemia Reperfusion Injury. Pharmaceuticals (Basel) 2022; 15:ph15060720. [PMID: 35745639 PMCID: PMC9228343 DOI: 10.3390/ph15060720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/02/2022] [Indexed: 11/23/2022] Open
Abstract
Tirzepatide is a dual glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptor agonist and a promising therapy for type 2 diabetes mellitus (T2DM). GLP-1 is an incretin hormone with therapeutic potential beyond type 2 diabetes mellitus. However, GLP-1 is rapidly degraded by dipeptdyl peptidase-IV (DPP-IV) to GLP-1 (9-36). Exendin-4 (Ex-4) is a DPP-IV-resistant GLP-1 receptor agonist which, when truncated to Ex-4 (9-39), acts as a GLP-1 receptor antagonist. In the present study, hearts isolated from Wistar rats (n = 8 per group) were perfused with a modified Langendorff preparation. Left ventricular (LV) contractility and cardiovascular hemodynamics were evaluated by a data acquisition program and infarct size was evaluated by 2,3,5-Triphenyl-2H-tetrazolium chloride (TTC) staining and cardiac enzyme levels. Hearts were subjected to 30 min regional ischemia, produced by ligation of the left anterior descending (LAD) coronary artery followed by 30 min reperfusion. Hearts were treated during reperfusion with either the non-lipidated precursor of tirzepatide (NLT), GLP-1, GLP-1 (9-36), or Ex-4 in the presence or absence of Ex-4 (9-39). Infusion of GLP-1 (9-36) or Ex-4 protected the heart against I/R injury (p > 0.01) by normalizing cardiac hemodynamic and enzyme levels. Neither GLP-1, NLT, nor Ex-4 (9-39) showed any protection. Interestingly, Ex-4 (9-39) blocked Ex-4-mediated protection but not that of GLP-1 (9-36). These data suggest that Ex-4-mediated protection is GLP-1-receptor-dependent but GLP-1 (9-36)-mediated protection is not.
Collapse
|
97
|
Ghorbanzadeh V, Jafarpour A, Pirnia A, Pajouhi N, Khaksarian M, Veiskarami S, Nazari A. The role of vasopressin V1A and oxytocin OTR receptors in protective effects of arginine vasopressin against H 2O 2-induced oxidative stress in H9C2 cells. Arch Physiol Biochem 2022; 128:830-835. [PMID: 32141340 DOI: 10.1080/13813455.2020.1729816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Oxidative stress, has been shown to play an important role in the pathophysiology of cardiac remodelling and heart failure. The aim of study is effect of arginine vasopressin (AVP) on apoptosis of cardiomyocyte via its receptors. MATERIALS AND METHODS The cell viability effect of AVP in H9C2 cardiomyocytes was assayed using the MTT method. The transcription and translation level of apoptosis genes (Bax, Bcl-2, caspase-3) were discovered with qRT-PCR and western blotting. RESULTS The results showed that vasopressin could reduce apoptosis in cardiomyocytes cell line through downregulation of caspase-3, BAX and upregulation of Bcl-2 (p < .001). Also, there was a decrease in anti-apoptosis effect of vasopressin when V1A and OTR receptors were blocked with their antagonists. DISCUSSION These results suggest that activation of V1A and OTR receptors in H9C2 cells mediate protective effect of vasopressin via regulating apoptosis marker that lead to cell survival under conditions of stress oxidative.Key pointAVP may contribute to the improvement of heart ischaemia through its actions on V1A and OTR receptors.
Collapse
Affiliation(s)
- Vajihe Ghorbanzadeh
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
| | - Afsaneh Jafarpour
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Afshin Pirnia
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
| | - Naser Pajouhi
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
- Department of Physiology, Lorestan University of Medical Science, Khorramabad, Iran
| | - Mojtaba Khaksarian
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
- Department of Physiology, Lorestan University of Medical Science, Khorramabad, Iran
| | - Saeed Veiskarami
- Department of animal science, Lorestan Agricultural and Natural Resources Research and Education Center, Khorramabad, Iran
| | - Afshin Nazari
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences Khorramabad, Iran
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
- Department of Physiology, Lorestan University of Medical Science, Khorramabad, Iran
| |
Collapse
|
98
|
Shiwakoti S, Ko JY, Gong D, Dhakal B, Lee JH, Adhikari R, Gwak Y, Park SH, Jun Choi I, Schini-Kerth VB, Kang KW, Oak MH. Effects of polystyrene nanoplastics on endothelium senescence and its underlying mechanism. ENVIRONMENT INTERNATIONAL 2022; 164:107248. [PMID: 35461096 DOI: 10.1016/j.envint.2022.107248] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/03/2022] [Accepted: 04/13/2022] [Indexed: 06/14/2023]
Abstract
Global plastic use has increased rapidly, and environmental pollution associated with nanoplastics (NPs) has been a growing concern recently. However, the impact and biological mechanism of NPs on the cardiovascular system are not well characterized. This study aimed to assess the possibility that NPs exposure promotes premature endothelial cell (EC) senescence in porcine coronary artery ECs and, if so, to elucidate the underlying mechanism. Treatment of ECs with NPs promoted the acquisition of senescence markers, senescence-associated β-galactosidase activity, and p53, p21, and p16 protein expression, resulting in the inhibition of proliferation. In addition, NPs impaired endothelium-dependent vasorelaxation associated with decreased endothelial nitric oxide synthase (eNOS) expression. NPs enhanced reactive oxygen species formation in ECs, and increased oxidative stress levels were associated with the induction of NADPH oxidases expression, followed by the subsequent downregulation of Sirt1 expression. The characteristics of EC senescence and dysfunction caused by NPs are prevented by an antioxidant (N-acetylcysteine), an NADPH oxidase inhibitor (apocynin), and a Sirt1 activator (resveratrol). These findings indicate that NPs induced premature EC senescence, at least in part, through the redox-sensitive eNOS/Sirt1 signaling pathway. This study suggested the effects and underlying mechanism of NPs on the cardiovascular system, which may provide pharmacological targets to prevent NPs-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Saugat Shiwakoti
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Ju-Young Ko
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Dalseong Gong
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea; Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260 INSERM (French National Institute of Health and Medical Research), University of Strasbourg, Strasbourg, France
| | - Bikalpa Dhakal
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Jeong-Hye Lee
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Radhika Adhikari
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Yeonhyang Gwak
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Sin-Hee Park
- Catholic Research Institute for Intractable Cardiovascular Disease (CRID), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ik Jun Choi
- Catholic Research Institute for Intractable Cardiovascular Disease (CRID), College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Valérie B Schini-Kerth
- Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260 INSERM (French National Institute of Health and Medical Research), University of Strasbourg, Strasbourg, France
| | - Ki-Woon Kang
- Division of Cardiology, Cardiovascular and Arrhythmia Center, Chung-Ang University, Seoul, Republic of Korea
| | - Min-Ho Oak
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, Republic of Korea.
| |
Collapse
|
99
|
Munjral S, Maindarkar M, Ahluwalia P, Puvvula A, Jamthikar A, Jujaray T, Suri N, Paul S, Pathak R, Saba L, Chalakkal RJ, Gupta S, Faa G, Singh IM, Chadha PS, Turk M, Johri AM, Khanna NN, Viskovic K, Mavrogeni S, Laird JR, Pareek G, Miner M, Sobel DW, Balestrieri A, Sfikakis PP, Tsoulfas G, Protogerou A, Misra DP, Agarwal V, Kitas GD, Kolluri R, Teji J, Al-Maini M, Dhanjil SK, Sockalingam M, Saxena A, Sharma A, Rathore V, Fatemi M, Alizad A, Viswanathan V, Krishnan PR, Omerzu T, Naidu S, Nicolaides A, Fouda MM, Suri JS. Cardiovascular Risk Stratification in Diabetic Retinopathy via Atherosclerotic Pathway in COVID-19/Non-COVID-19 Frameworks Using Artificial Intelligence Paradigm: A Narrative Review. Diagnostics (Basel) 2022; 12:1234. [PMID: 35626389 PMCID: PMC9140106 DOI: 10.3390/diagnostics12051234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetes is one of the main causes of the rising cases of blindness in adults. This microvascular complication of diabetes is termed diabetic retinopathy (DR) and is associated with an expanding risk of cardiovascular events in diabetes patients. DR, in its various forms, is seen to be a powerful indicator of atherosclerosis. Further, the macrovascular complication of diabetes leads to coronary artery disease (CAD). Thus, the timely identification of cardiovascular disease (CVD) complications in DR patients is of utmost importance. Since CAD risk assessment is expensive for low-income countries, it is important to look for surrogate biomarkers for risk stratification of CVD in DR patients. Due to the common genetic makeup between the coronary and carotid arteries, low-cost, high-resolution imaging such as carotid B-mode ultrasound (US) can be used for arterial tissue characterization and risk stratification in DR patients. The advent of artificial intelligence (AI) techniques has facilitated the handling of large cohorts in a big data framework to identify atherosclerotic plaque features in arterial ultrasound. This enables timely CVD risk assessment and risk stratification of patients with DR. Thus, this review focuses on understanding the pathophysiology of DR, retinal and CAD imaging, the role of surrogate markers for CVD, and finally, the CVD risk stratification of DR patients. The review shows a step-by-step cyclic activity of how diabetes and atherosclerotic disease cause DR, leading to the worsening of CVD. We propose a solution to how AI can help in the identification of CVD risk. Lastly, we analyze the role of DR/CVD in the COVID-19 framework.
Collapse
Affiliation(s)
- Smiksha Munjral
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (S.M.); (M.M.); (A.P.); (A.J.); (T.J.); (I.M.S.); (P.S.C.); (S.K.D.)
| | - Mahesh Maindarkar
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (S.M.); (M.M.); (A.P.); (A.J.); (T.J.); (I.M.S.); (P.S.C.); (S.K.D.)
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India;
| | - Puneet Ahluwalia
- Max Institute of Cancer Care, Max Super Specialty Hospital, New Delhi 110017, India;
| | - Anudeep Puvvula
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (S.M.); (M.M.); (A.P.); (A.J.); (T.J.); (I.M.S.); (P.S.C.); (S.K.D.)
- Annu’s Hospitals for Skin and Diabetes, Nellore 524101, India
| | - Ankush Jamthikar
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (S.M.); (M.M.); (A.P.); (A.J.); (T.J.); (I.M.S.); (P.S.C.); (S.K.D.)
| | - Tanay Jujaray
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (S.M.); (M.M.); (A.P.); (A.J.); (T.J.); (I.M.S.); (P.S.C.); (S.K.D.)
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95616, USA
| | - Neha Suri
- Mira Loma High School, Sacramento, CA 95821, USA;
| | - Sudip Paul
- Department of Biomedical Engineering, North Eastern Hill University, Shillong 793022, India;
| | - Rajesh Pathak
- Department of Computer Science Engineering, Rawatpura Sarkar University, Raipur 492015, India;
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, 40138 Cagliari, Italy; (L.S.); (A.B.)
| | | | - Suneet Gupta
- CSE Department, Bennett University, Greater Noida 201310, India;
| | - Gavino Faa
- Department of Pathology, Azienda Ospedaliero Universitaria, 09124 Cagliari, Italy;
| | - Inder M. Singh
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (S.M.); (M.M.); (A.P.); (A.J.); (T.J.); (I.M.S.); (P.S.C.); (S.K.D.)
| | - Paramjit S. Chadha
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (S.M.); (M.M.); (A.P.); (A.J.); (T.J.); (I.M.S.); (P.S.C.); (S.K.D.)
| | - Monika Turk
- The Hanse-Wissenschaftskolleg Institute for Advanced Study, 27753 Delmenhorst, Germany;
| | - Amer M. Johri
- Department of Medicine, Division of Cardiology, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Narendra N. Khanna
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi 110001, India; (N.N.K.); (A.S.)
| | - Klaudija Viskovic
- Department of Radiology and Ultrasound, University Hospital for Infectious Diseases, 10 000 Zagreb, Croatia;
| | - Sophie Mavrogeni
- Cardiology Clinic, Onassis Cardiac Surgery Centre, 17674 Athens, Greece;
| | - John R. Laird
- Heart and Vascular Institute, Adventist Health St. Helena, St. Helena, CA 94574, USA;
| | - Gyan Pareek
- Minimally Invasive Urology Institute, Brown University, Providence, RI 02912, USA;
| | - Martin Miner
- Men’s Health Centre, Miriam Hospital Providence, Providence, RI 02906, USA;
| | - David W. Sobel
- Rheumatology Unit, National Kapodistrian University of Athens, 15772 Athens, Greece; (D.W.S.); (P.P.S.)
| | - Antonella Balestrieri
- Department of Radiology, Azienda Ospedaliero Universitaria, 40138 Cagliari, Italy; (L.S.); (A.B.)
| | - Petros P. Sfikakis
- Rheumatology Unit, National Kapodistrian University of Athens, 15772 Athens, Greece; (D.W.S.); (P.P.S.)
| | - George Tsoulfas
- Department of Surgery, Aristoteleion University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Athanasios Protogerou
- Cardiovascular Prevention and Research Unit, Department of Pathophysiology, National & Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Durga Prasanna Misra
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India; (D.P.M.); (V.A.)
| | - Vikas Agarwal
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India; (D.P.M.); (V.A.)
| | - George D. Kitas
- Academic Affairs, Dudley Group NHS Foundation Trust, Dudley DY1 2HQ, UK;
- Arthritis Research UK Epidemiology Unit, Manchester University, Manchester M13 9PL, UK
| | - Raghu Kolluri
- OhioHealth Heart and Vascular, Columbus, OH 43214, USA;
| | - Jagjit Teji
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA;
| | - Mustafa Al-Maini
- Allergy, Clinical Immunology and Rheumatology Institute, Toronto, ON L4Z 4C4, Canada;
| | - Surinder K. Dhanjil
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (S.M.); (M.M.); (A.P.); (A.J.); (T.J.); (I.M.S.); (P.S.C.); (S.K.D.)
| | | | - Ajit Saxena
- Department of Cardiology, Indraprastha APOLLO Hospitals, New Delhi 110001, India; (N.N.K.); (A.S.)
| | - Aditya Sharma
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA 22904, USA;
| | - Vijay Rathore
- Nephrology Department, Kaiser Permanente, Sacramento, CA 95119, USA;
| | - Mostafa Fatemi
- Department of Physiology & Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA;
| | - Azra Alizad
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA;
| | - Vijay Viswanathan
- MV Hospital for Diabetes and Professor MVD Research Centre, Chennai 600013, India;
| | | | - Tomaz Omerzu
- Department of Neurology, University Medical Centre Maribor, 1262 Maribor, Slovenia;
| | - Subbaram Naidu
- Electrical Engineering Department, University of Minnesota, Duluth, MN 55812, USA;
| | - Andrew Nicolaides
- Vascular Screening and Diagnostic Centre, University of Nicosia Medical School, Nicosia 2408, Cyprus;
| | - Mostafa M. Fouda
- Department of Electrical and Computer Engineering, Idaho State University, Pocatello, ID 83209, USA;
| | - Jasjit S. Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA 95661, USA; (S.M.); (M.M.); (A.P.); (A.J.); (T.J.); (I.M.S.); (P.S.C.); (S.K.D.)
| |
Collapse
|
100
|
Greenberg HZE, Zhao G, Shah AM, Zhang M. Role of oxidative stress in calcific aortic valve disease and its therapeutic implications. Cardiovasc Res 2022; 118:1433-1451. [PMID: 33881501 PMCID: PMC9074995 DOI: 10.1093/cvr/cvab142] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the end result of active cellular processes that lead to the progressive fibrosis and calcification of aortic valve leaflets. In western populations, CAVD is a significant cause of cardiovascular morbidity and mortality, and in the absence of effective drugs, it will likely represent an increasing disease burden as populations age. As there are currently no pharmacological therapies available for preventing, treating, or slowing the development of CAVD, understanding the mechanisms underlying the initiation and progression of the disease is important for identifying novel therapeutic targets. Recent evidence has emerged of an important causative role for reactive oxygen species (ROS)-mediated oxidative stress in the pathophysiology of CAVD, inducing the differentiation of valve interstitial cells into myofibroblasts and then osteoblasts. In this review, we focus on the roles and sources of ROS driving CAVD and consider their potential as novel therapeutic targets for this debilitating condition.
Collapse
Affiliation(s)
- Harry Z E Greenberg
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Guoan Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Henan, China
| | - Ajay M Shah
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Min Zhang
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|