151
|
PTEN mediates serum deprivation-induced cytotoxicity in H9c2 cells via the PI3K/AKT signaling pathway. Toxicol In Vitro 2021; 73:105131. [PMID: 33652126 DOI: 10.1016/j.tiv.2021.105131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/09/2021] [Accepted: 02/25/2021] [Indexed: 12/22/2022]
Abstract
The pathogenesis of acute myocardial infarction (AMI) is associated with cardiomyocyte necrosis and apoptosis. Numerous studies have determined the regulatory effects of Phosphatase and tensin homolog (PTEN) cell proliferation and apoptosis in other cell types. However, the potential role of PTEN in cardiomyocyte is unclear. In this study, we used H9c2 cells cultured under serum deprivation to simulate the apoptosis process of myocardial infarction. Small interference RNA (siRNA) of PTEN was used to knock down the expression of PTEN. Cell viability was determined by CCK-8. Cell proliferation was examined by Edu staining, and the protein expression was analyzed by Western blot. We also evaluated the generation of ROS, the degree of DNA damage, and cell apoptosis using immunofluorescence assay. As a result, we observed that serum deprivation in H9c2 cells increased PTEN expression. Functionally, the PTEN knockdown experiment using siRNA inhibited serum deprivation-induced cell apoptosis, ROS production, and DNA damage, whereas increased cell proliferation. All these effects could be reversed by phosphatidylinositol 3-kinase (PI3K) inhibitor, which indicated the PI3K/protein kinase B (AKT) might be the critical component of the PTEN effects during serum deficiency. In conclusion, our study indicated the role of the PTEN/PI3K/AKT pathway in serum deprivation-induced cytotoxicity in H9c2 cells.
Collapse
|
152
|
Lomivorotov VV, Moroz G, Abubakirov M, Osinsky R, Landoni G. Volatile and Intravenous Anesthetics for Brain Protection in Cardiac Surgery: Does the Choice of Anesthesia Matter? J Cardiothorac Vasc Anesth 2021; 36:567-576. [PMID: 33766470 DOI: 10.1053/j.jvca.2021.02.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 11/11/2022]
Abstract
Postoperative neurologic complications have a significant effect on morbidity, mortality, and long-term disability in patients undergoing cardiac surgery. The etiology of brain injury in patients undergoing cardiac surgery is multifactorial and remains unclear. There are several perioperative causative factors for neurologic complications, including microembolization, hypoperfusion, and systemic inflammatory response syndrome. Despite technologic advances and the development of new anesthetic drugs, there remains a high rate of postoperative neurologic complications. Moreover, despite the strong evidence that volatile anesthesia exerts cardioprotective effects in patients undergoing cardiac surgery, the neuroprotective effects of volatile agents remain unclear. Several studies have reported an association of using volatile anesthetics with improvement of biochemical markers of brain injury and postoperative neurocognitive function. However, there is a need for additional studies to define the optimal anesthetic drug for protecting the brain in patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Vladimir V Lomivorotov
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia; Department of Anesthesiology and Intensive Care, Novosibirsk State University, Novosibirsk, Russia.
| | - Gleb Moroz
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Marat Abubakirov
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Roman Osinsky
- Department of Anesthesiology and Intensive Care, E. Meshalkin National Medical Research Center, Novosibirsk, Russia
| | - Giovanni Landoni
- Anesthesia and Intensive Care Department, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
153
|
Liu Y, Li M, Du X, Huang Z, Quan N. Sestrin 2, a potential star of antioxidant stress in cardiovascular diseases. Free Radic Biol Med 2021; 163:56-68. [PMID: 33310138 DOI: 10.1016/j.freeradbiomed.2020.11.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
Abstract
Physiological reactive oxygen species (ROS) play an important role in cellular signal transduction. However, excessive ROS is an important pathological mechanism in most cardiovascular diseases (CVDs), such as myocardial aging, cardiomyopathy, ischemia/reperfusion injury (e.g., myocardial infarction) and heart failure. Programmed cell death, hypertrophy and fibrosis may be due to oxidative stress. Sestrin 2 (Sesn2), a stress-inducible protein associated with various stress conditions, is a potential antioxidant. Sesn2 can suppress the process of heart damage caused by oxidative stress, promote cell survival and play a key role in a variety of CVDs. This review discusses the effect of Sesn2 on the redox signal, mainly via participation in the signaling pathway of nuclear factor erythroid 2-related factor 2, activation of adenosine monophosphate-activated protein kinase and inhibition of mammalian target of rapamycin complex 1. It also discusses the effect of Sesn2's antioxidant activity on different CVDs. We speculate that Sesn2 plays an important role in CVDs by stimulating the process of antioxidation and promoting the adaptation of cells to stress conditions and/or the environment, opening a new avenue for related therapeutic strategies.
Collapse
Affiliation(s)
- Yunxia Liu
- Department of Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Meina Li
- Department of Infection Control, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xiaoyu Du
- Department of Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Zhehao Huang
- Department of Neurosurgery, The Third Hospital of Jilin University, Changchun, Jilin, 130031, China.
| | - Nanhu Quan
- Department of Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
154
|
Carrasco R, Castillo RL, Gormaz JG, Carrillo M, Thavendiranathan P. Role of Oxidative Stress in the Mechanisms of Anthracycline-Induced Cardiotoxicity: Effects of Preventive Strategies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8863789. [PMID: 33574985 PMCID: PMC7857913 DOI: 10.1155/2021/8863789] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/29/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022]
Abstract
Anthracycline-induced cardiotoxicity (AIC) persists as a significant cause of morbidity and mortality in cancer survivors. Although many protective strategies have been evaluated, cardiotoxicity remains an ongoing threat. The mechanisms of AIC remain unclear; however, several pathways have been proposed, suggesting a multifactorial origin. When the central role of topoisomerase 2β in the pathophysiology of AIC was described some years ago, the classical reactive oxygen species (ROS) hypothesis shifted to a secondary position. However, new insights have reemphasized the importance of the role of oxidative stress-mediated signaling as a common pathway and a critical modulator of the different mechanisms involved in AIC. A better understanding of the mechanisms of cardiotoxicity is crucial for the development of treatment strategies. It has been suggested that the available therapeutic interventions for AIC could act on the modulation of oxidative balance, leading to a reduction in oxidative stress injury. These indirect antioxidant effects make them an option for the primary prevention of AIC. In this review, our objective is to provide an update of the accumulated knowledge on the role of oxidative stress in AIC and the modulation of the redox balance by potential preventive strategies.
Collapse
Affiliation(s)
- Rodrigo Carrasco
- Division of Cardiology, Peter Munk Cardiac Centre and the Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| | - Rodrigo L. Castillo
- Medicine Department, East Division, Faculty of Medicine, University of Chile. Santiago, Chile; Critical Care Patient Unit, Hospital Salvador, Santiago, Chile
| | - Juan G. Gormaz
- Faculty of Medicine, University of Chile, Santiago, Chile
| | - Montserrat Carrillo
- Division of Cardiology, Peter Munk Cardiac Centre and the Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| | - Paaladinesh Thavendiranathan
- Division of Cardiology, Peter Munk Cardiac Centre and the Ted Rogers Centre for Heart Research, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
155
|
Barth syndrome: cardiolipin, cellular pathophysiology, management, and novel therapeutic targets. Mol Cell Biochem 2021; 476:1605-1629. [PMID: 33415565 DOI: 10.1007/s11010-020-04021-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022]
Abstract
Barth syndrome is a rare X-linked genetic disease classically characterized by cardiomyopathy, skeletal myopathy, growth retardation, neutropenia, and 3-methylglutaconic aciduria. It is caused by mutations in the tafazzin gene localized to chromosome Xq28.12. Mutations in tafazzin may result in alterations in the level and molecular composition of the mitochondrial phospholipid cardiolipin and result in large elevations in the lysophospholipid monolysocardiolipin. The increased monolysocardiolipin:cardiolipin ratio in blood is diagnostic for the disease, and it leads to disruption in mitochondrial bioenergetics. In this review, we discuss cardiolipin structure, synthesis, and function and provide an overview of the clinical and cellular pathophysiology of Barth Syndrome. We highlight known pharmacological management for treatment of the major pathological features associated with the disease. In addition, we discuss non-pharmacological management. Finally, we highlight the most recent promising therapeutic options for this rare mitochondrial disease including lipid replacement therapy, peroxisome proliferator-activated receptor agonists, tafazzin gene replacement therapy, induced pluripotent stem cells, mitochondria-targeted antioxidants and peptides, and the polyphenolic compound resveratrol.
Collapse
|
156
|
Liu Y, Zhang Y, Liu Q, Wang Q, Lin A, Luo J, Du Y, Lin YW, Wei H. In vitro measurement of superoxide dismutase-like nanozyme activity: a comparative study. Analyst 2021; 146:1872-1879. [DOI: 10.1039/d0an02164d] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Analyzing the SOD-like activity of nanozymes in vitro is of great importance for identifying new nanozymes and predicting their potential biological effects in vivo.
Collapse
Affiliation(s)
- Yufeng Liu
- Department of Biomedical Engineering
- College of Engineering and Applied Sciences
- Nanjing University
- Nanjing
- China
| | - Yihong Zhang
- Department of Biomedical Engineering
- College of Engineering and Applied Sciences
- Nanjing University
- Nanjing
- China
| | - Quanyi Liu
- State Key Laboratory of Electroanalytical Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Science
- Changchun
- China
| | - Quan Wang
- Department of Biomedical Engineering
- College of Engineering and Applied Sciences
- Nanjing University
- Nanjing
- China
| | - Anqi Lin
- Department of Biomedical Engineering
- College of Engineering and Applied Sciences
- Nanjing University
- Nanjing
- China
| | - Jie Luo
- Laboratory of Protein Structure and Function
- School of Chemistry and Chemical Engineering
- University of South China
- Hengyang
- China
| | - Yan Du
- State Key Laboratory of Electroanalytical Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Science
- Changchun
- China
| | - Ying-Wu Lin
- Laboratory of Protein Structure and Function
- School of Chemistry and Chemical Engineering
- University of South China
- Hengyang
- China
| | - Hui Wei
- Department of Biomedical Engineering
- College of Engineering and Applied Sciences
- Nanjing University
- Nanjing
- China
| |
Collapse
|
157
|
Scheen M, Giraud R, Bendjelid K. Stress hyperglycemia, cardiac glucotoxicity, and critically ill patient outcomes current clinical and pathophysiological evidence. Physiol Rep 2021; 9:e14713. [PMID: 33463901 PMCID: PMC7814494 DOI: 10.14814/phy2.14713] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 01/07/2023] Open
Abstract
Stress hyperglycemia is a transient increase in blood glucose during acute physiological stress in the absence of glucose homeostasis dysfunction. Its's presence has been described in critically ill patients who are subject to many physiological insults. In this regard, hyperglycemia and impaired glucose tolerance are also frequent in patients who are admitted to the intensive care unit for heart failure and cardiogenic shock. The hyperglycemia observed at the beginning of these cardiac disorders appears to be related to a variety of stress mechanisms. The release of major stress and steroid hormones, catecholamine overload, and glucagon all participate in generating a state of insulin resistance with increased hepatic glucose output and glycogen breakdown. In fact, the observed pathophysiological response, which appears to regulate a stress situation, is harmful because it induces mitochondrial impairment, oxidative stress-related injury to cells, endothelial damage, and dysfunction of several cellular channels. Paradigms are now being challenged by growing evidence of a phenomenon called glucotoxicity, providing an explanation for the benefits of lowering glucose levels with insulin therapy in these patients. In the present review, the authors present the data published on cardiac glucotoxicity and discuss the benefits of lowering plasma glucose to improve heart function and to positively affect the course of critical illness.
Collapse
Affiliation(s)
- Marc Scheen
- Intensive Care DivisionUniversity HospitalsGenevaSwitzerland
- Geneva Hemodynamic Research GroupGenevaSwitzerland
- Faculty of MedicineGenevaSwitzerland
| | - Raphael Giraud
- Intensive Care DivisionUniversity HospitalsGenevaSwitzerland
- Geneva Hemodynamic Research GroupGenevaSwitzerland
- Faculty of MedicineGenevaSwitzerland
| | - Karim Bendjelid
- Intensive Care DivisionUniversity HospitalsGenevaSwitzerland
- Geneva Hemodynamic Research GroupGenevaSwitzerland
- Faculty of MedicineGenevaSwitzerland
| |
Collapse
|
158
|
MIYAZAWA T. Lipid hydroperoxides in nutrition, health, and diseases. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2021; 97:161-196. [PMID: 33840675 PMCID: PMC8062262 DOI: 10.2183/pjab.97.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/08/2021] [Indexed: 05/08/2023]
Abstract
Research on lipid peroxidation in food degradation, oil and fat nutrition, and age-related diseases has gained significant international attention for the view of improvement of societal health and longevity. In order to promote basic studies on these topics, a chemiluminescence detection-high performance liquid chromatography instrument using a high-sensitivity single photon counter as a detector was developed. This instrument enabled us to selectively detect and quantify lipid hydroperoxides, a primary product of lipid peroxidation reactions, as hydroperoxide groups at the lipid class level. Furthermore, an analytical method using liquid chromatography-tandem mass spectrometry has been established to discriminate the position and stereoisomerization of hydroperoxide groups in lipid hydroperoxides. Using these two methods, the reaction mechanisms of lipid peroxidation in food and in the body have been confirmed.
Collapse
Affiliation(s)
- Teruo MIYAZAWA
- Food Biotechnology Platform Promoting Project, New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
159
|
Liu C, Yan Q, Gao C, Lin L, Wei J. Study on antioxidant effect of recombinant glutathione peroxidase 1. Int J Biol Macromol 2020; 170:503-513. [PMID: 33383079 DOI: 10.1016/j.ijbiomac.2020.12.183] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/19/2022]
Abstract
Glutathione peroxidase 1 (GPx1) is an important antioxidant selenium enzyme and has a good prospect for drug development. However, the expression of GPx1 requires a complex expression mechanism, which makes the drug development of recombinant GPx1 (rGPx1) difficult. In the previous study, we expressed highly active rhGPx1 in amber-less Escherichia coli by using a novel chimeric tRNAUTuT6. However, the antioxidant effect of rhGPx1 at the cellular and animal levels has not been verified. In this study, we established isoproterenol (ISO)-induced oxidative stress injury models to study the antioxidant effect of rhGPx1 at the cellular and animal levels. Meanwhile, in order to more accurately reflect the antioxidant effect of rGPx1 in mice, we used the same method to express recombinant mouse GPx1 (rmGPx1) as a control for rhGPx1. The results of a study showed that rhGPx1 has a good antioxidant effect at the cellular and animal levels. However, due to species differences, rhGPx1 had immunogenicity in mice and antibodies of rhGPx1 could inhibit its antioxidant activity, so the antioxidant effect of rhGPx1 was not as good as rmGPx1 in mice. Nevertheless, this study provides a reliable theoretical basis for the development of rhGPx1 as an antioxidant drug.
Collapse
Affiliation(s)
- Chang Liu
- College of Pharmaceutical Science, Jilin University, Changchun 130021, PR China
| | - Qi Yan
- College of Pharmaceutical Science, Jilin University, Changchun 130021, PR China
| | - Chao Gao
- College of Pharmaceutical Science, Jilin University, Changchun 130021, PR China
| | - Liangru Lin
- College of Pharmaceutical Science, Jilin University, Changchun 130021, PR China
| | - Jingyan Wei
- College of Pharmaceutical Science, Jilin University, Changchun 130021, PR China; Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun 130000, PR China.
| |
Collapse
|
160
|
Li W, Wang Y, Wei H, Zhang Y, Guo Z, Qiu Y, Wen L, Xie Z. Structural characterization of Lanzhou lily (Lilium davidii var. unicolor) polysaccharides and determination of their associated antioxidant activity. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:5603-5616. [PMID: 32608519 DOI: 10.1002/jsfa.10613] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 05/25/2020] [Accepted: 07/01/2020] [Indexed: 05/27/2023]
Abstract
BACKGROUD The Lanzhou lily (Lilium davidii var. unicolor) is the only Lilium species that is used for both culinary and medicinal purposes in China. Its bulbs contain various bioactive substances, such as polysaccharides, saponins and colchicine. Lanzhou lily polysaccharides are known to have anti-immunity, anti-tumor and anti-oxidation functions. RESULTS The present study used a Box-Behnken design to optimize the ultrasound-assisted extraction of Lanzhou lily polysaccharides. Compared to other enzymes, trypsin significantly increased the polysaccharide yields, whereas the protein content of polysaccharides extracted with trypsin was the lowest. Monosaccharide mainly includes glucose (> 50%) and mannose (> 10%). 1,1-Diphenyl-2-picrylhydrazyl radical scavenging activity, chelating activity, total antioxidant capacity and hydroxyl radical scavenging activity of Lanzhou lily polysaccharides extracted with trypsin were stronger than those extracted without enzymes (control). Structural characteristics of Lanzhou lily polysaccharides extracted with trypsin and extracted without enzymes were characterized by scanning electron microscopy and nuclear magnetic resonance spectroscopy. When water extracted polysaccharide and trypsin extracted polysaccharide concentrations were 200 μg mL-1 , Raw264.7 proliferation rates were 101.69% and 159.41%, respectively. CONCLUSION The Lanzhou lily polysaccharide was identified as α-(1 → 6)-d-glucan. Consequently, the effects of both potential antioxidant and proliferative activity of trypsin are significant. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wenmei Li
- Northwest Institute of Eco-environment and Resource, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yajun Wang
- Northwest Institute of Eco-environment and Resource, Chinese Academy of Sciences, Lanzhou, China
| | - Hailian Wei
- Northwest Institute of Eco-environment and Resource, Chinese Academy of Sciences, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yubao Zhang
- Northwest Institute of Eco-environment and Resource, Chinese Academy of Sciences, Lanzhou, China
| | - Zhihong Guo
- Northwest Institute of Eco-environment and Resource, Chinese Academy of Sciences, Lanzhou, China
| | - Yang Qiu
- Northwest Institute of Eco-environment and Resource, Chinese Academy of Sciences, Lanzhou, China
| | - Lingrong Wen
- Ministry of Agriculture, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Zhongkui Xie
- Northwest Institute of Eco-environment and Resource, Chinese Academy of Sciences, Lanzhou, China
| |
Collapse
|
161
|
Ertelt A, Stumpff F, Merle R, Kuban S, Bollinger L, Liertz S, Gehlen H. Asymmetric dimethylarginine-A potential cardiac biomarker in horses. J Vet Cardiol 2020; 33:43-51. [PMID: 33360109 DOI: 10.1016/j.jvc.2020.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 11/06/2020] [Accepted: 11/17/2020] [Indexed: 12/22/2022]
Abstract
INTRODUCTION/OBJECTIVES Asymmetric dimethylarginine (ADMA) is a cardiac biomarker in humans, symmetric dimethylarginine (SDMA) a renal biomarker in humans, cats, and dogs. The purpose of this prospective study was to investigate if measuring serum ADMA and SDMA concentrations via ELISA allows detection of cardiac disease in horses in a routine laboratory setting. In this context, reference values in horses were established. ANIMALS, MATERIALS, AND METHODS Seventy-eight horses with no known medical history were compared to 23 horses with confirmed structural cardiac disease with/or without arrhythmias. Horses underwent physical examination, electrocardiography, echocardiography and venous blood sampling and were staged based on the severity of cardiac disease from 0 to II. Asymmetric dimethylarginine and SDMA were measured via ELISA and crosschecked using liquid chromatograph triple quadrupole mass spectrometry. Reference intervals with 90th percent confidence intervals were evaluated and standard software was used to test for significant differences in ADMA, SDMA, and the l-arginine/ADMA ratio between groups. RESULTS The reference ranges were 1.7-3.8 μmol/L and 0.3-0.8 μmol/L for ADMA and SDMA, respectively. Serum ADMA was higher in horses with heart disease compared to healthy horses (p < 0.01) and highest in horses with stage II heart disease (p = 0.02). The l-Arginine/ADMA ratio was significantly higher in healthy animals than those with cardiac disease (p = 0.001). CONCLUSIONS Reference values for serum ADMA and SDMA using ELISA methods are presented in horses. This study confirms the association between heart disease and increased serum ADMA concentration as well as a decreased l-Arginine/ADMA ratio in horses.
Collapse
Affiliation(s)
- A Ertelt
- Equine Clinic, Internal Medicine, Freie Universitaet Berlin, Oertzenweg 19b, 14193, Berlin, Germany.
| | - F Stumpff
- Institute of Veterinary Physiology, Freie Universitaet Berlin, Oertzenweg 19b, 14163, Berlin, Germany
| | - R Merle
- Institute for Veterinary Epidemiology and Biostatistics, Freie Universitaet Berlin, Koenigsweg 67, 14163, Berlin, Germany
| | - S Kuban
- Equine Clinic, Internal Medicine, Freie Universitaet Berlin, Oertzenweg 19b, 14193, Berlin, Germany
| | - L Bollinger
- Equine Clinic, Internal Medicine, Freie Universitaet Berlin, Oertzenweg 19b, 14193, Berlin, Germany
| | - S Liertz
- Equine Clinic, Internal Medicine, Freie Universitaet Berlin, Oertzenweg 19b, 14193, Berlin, Germany
| | - H Gehlen
- Equine Clinic, Internal Medicine, Freie Universitaet Berlin, Oertzenweg 19b, 14193, Berlin, Germany
| |
Collapse
|
162
|
Nwasike C, Yoo E, Purr E, Doiron AL. Activatable superparamagnetic iron oxide nanoparticles scavenge reactive oxygen species in macrophages and endothelial cells. RSC Adv 2020; 10:41305-41314. [PMID: 35516581 PMCID: PMC9057763 DOI: 10.1039/d0ra06683d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/05/2020] [Indexed: 01/11/2023] Open
Abstract
Reactive oxygen species (ROS) are key markers of inflammation, with varying levels of superoxide indicating the degree of inflammation. Inflammatory diseases remain the leading cause of death in the developed world. Previously, we showed that interpolymer complexed superparamagnetic iron oxide nanoparticles (IPC-SPIOs) are capable of decomplexing and activating T2 magnetic resonance (MR) contrast in superoxide-rich environments. Here, we investigate the ability of IPC-SPIOs to scavenge ROS in immune and endothelial cells which should activate the superparamagnetic core. In exogenously generated superoxide, ROS scavenging by the nanoparticles was concentration dependent and ranged from 5% to over 50% of available ROS. A statistically significant reduction in ROS was observed in the presence of IPCSPIOs compared to poly(ethylene glycol)-coated SPIOs (PEG-SPIOs). During in vitro cellular assays, a reduction in ROS was observed in macrophages, monocytes, and human endothelial cells. Macrophages and endothelial cells experienced significantly higher ROS reduction compared to monocytes. ROS scavenging peaked 12 hours post-exposure to IPC-SPIOs in most studies, with some cell samples experiencing extended scavenging with increasing IPC-SPIO concentration. At the tested concentrations, particles were not cytotoxic, and confocal imaging showed localization of particles within cells. These findings demonstrate the potential of IPC-SPIOs as activatable MR contrast agents capable of activating under inflammation-induced cellular redox conditions as reporters of inflammatory disease severity or staging.
Collapse
Affiliation(s)
- Chukwuazam Nwasike
- Department of Biomedical Engineering, Binghamton University (SUNY) Binghamton NY 13902 USA
| | - Eunsoo Yoo
- Department of Biomedical Engineering, Binghamton University (SUNY) Binghamton NY 13902 USA
| | - Erin Purr
- Department of Biomedical Engineering, Binghamton University (SUNY) Binghamton NY 13902 USA
| | - Amber L Doiron
- Department of Electrical and Biomedical Engineering, University of Vermont Burlington VT 05405 USA +1-802-656-3382
| |
Collapse
|
163
|
Oxidation of ethidium-based probes by biological radicals: mechanism, kinetics and implications for the detection of superoxide. Sci Rep 2020; 10:18626. [PMID: 33122809 PMCID: PMC7596101 DOI: 10.1038/s41598-020-75373-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/13/2020] [Indexed: 12/21/2022] Open
Abstract
Hydroethidine (HE) and hydropropidine (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {HPr}^{+}$$\end{document}HPr+) are fluorogenic probes used for the detection of the intra- and extracellular superoxide radical anion (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {O}_{ {2}}^{\bullet -}$$\end{document}O2∙-). In this study, we provide evidence that HE and \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {HPr}^{+}$$\end{document}HPr+ react rapidly with the biologically relevant radicals, including the hydroxyl radical, peroxyl radicals, the trioxidocarbonate radical anion, nitrogen dioxide, and the glutathionyl radical, via one-electron oxidation, forming the corresponding radical cations. At physiological pH, the radical cations of the probes react rapidly with \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {O}_{ {2}}^{\bullet -}$$\end{document}O2∙-, leading to the specific 2-hydroxylated cationic products. We determined the rate constants of the reaction between \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {O}_{ {2}}^{\bullet -}$$\end{document}O2∙- and the radical cations of the probes. We also synthesized N-methylated analogs of \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {HPr}^{+}$$\end{document}HPr+ and HE which were used in mechanistic studies. Methylation of the amine groups was not found to prevent the reaction between the radical cation of the probe and the superoxide, but it significantly increased the lifetime of the radical cation and had a substantial effect on the profiles of the oxidation products by inhibiting the formation of dimeric products. We conclude that the N-methylated analogs of HE and \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {HPr}^{+}$$\end{document}HPr+ may be used as a scaffold for the design of a new generation of probes for intra- and extracellular superoxide.
Collapse
|
164
|
A systematic review of post-translational modifications in the mitochondrial permeability transition pore complex associated with cardiac diseases. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165992. [PMID: 33091565 DOI: 10.1016/j.bbadis.2020.165992] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/08/2020] [Indexed: 12/28/2022]
Abstract
The mitochondrial permeability transition pore (mPTP) opening is involved in the pathophysiology of multiple cardiac diseases, such as ischemia/reperfusion injury and heart failure. A growing number of evidence provided by proteomic screening techniques has demonstrated the role of post-translational modifications (PTMs) in several key components of the pore in response to changes in the extra/intracellular environment and bioenergetic demand. This could lead to a fine, complex regulatory mechanism that, under pathological conditions, can shift the state of mitochondrial functions and, thus, the cell's fate. Understanding the complex relationship between these PTMs is still under investigation and can provide new, promising therapeutic targets and treatment approaches. This review, using a systematic review of the literature, presents the current knowledge on PTMs of the mPTP and their role in health and cardiac disease.
Collapse
|
165
|
Gouda E, Babiker F. Micronized flavonoid fraction Daflon 500 protects heart against ischemia–reperfusion injury: an old medicine for a new target. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1832921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Eman Gouda
- Department of Physiology, Faculty of Medicine, Health Science Center, Kuwait University, Kuwait, Kuwait
| | - Fawzi Babiker
- Department of Physiology, Faculty of Medicine, Health Science Center, Kuwait University, Kuwait, Kuwait
| |
Collapse
|
166
|
Liu N, Li Y, Nan W, Zhou W, Huang J, Li R, Zhou L, Hu R. Interaction of TPPP3 with VDAC1 Promotes Endothelial Injury through Activation of Reactive Oxygen Species. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5950195. [PMID: 33082910 PMCID: PMC7556057 DOI: 10.1155/2020/5950195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/27/2020] [Accepted: 09/07/2020] [Indexed: 11/22/2022]
Abstract
Endothelial injury plays a critical role in the pathogenesis of cardiovascular disorders and metabolic-associated vascular complications which are the leading cause of death worldwide. However, the mechanism underlying endothelial dysfunction is not completely understood. The study is aimed at investigating the role of tubulin polymerization-promoting protein family member 3 (TPPP3) in palmitic acid- (PA-) induced endothelial injury. The effect of TPPP3 on human umbilical vein endothelial cells (HUVECs) was determined by evaluating apoptosis, tube formation, and reactive oxygen species (ROS) production. TPPP3 silencing inhibited PA overload-induced apoptosis and production of ROS, along with the alteration of apoptosis-related key proteins such as BCL-2 and Bax. Mechanically, voltage-dependent anion channel 1 (VDAC1) was identified as a novel functional binding partner of TPPP3, and TPPP3 promoted VDAC1 protein stability and its activity. Further studies indicated that TPPP3 could promote apoptosis, ROS production, tube formation, and proapoptotic protein expression and reduce antiapoptotic protein expression through increasing VDAC1 expression under mildly elevated levels of PA. Collectively, these results demonstrated that TPPP3 could promote PA-induced oxidative damage in HUVECs via a VDAC1-dependent pathway, suggesting that TPPP3 might be considered as a potential therapeutic target in vascular disease.
Collapse
Affiliation(s)
- Naijia Liu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yintao Li
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wu Nan
- Department of Geriatrics, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenbai Zhou
- Department of Medicine, Emanuel Medical Center, Turlock, California, USA
| | - Jinya Huang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Rumei Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Linuo Zhou
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Renming Hu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, China
| |
Collapse
|
167
|
Zhang XW, Zhou M, An L, Zhang P, Li P, Chen J. Lipophilic Extract and Tanshinone IIA Derived from Salvia miltiorrhiza Attenuate Uric Acid Nephropathy through Suppressing Oxidative Stress-Activated MAPK Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1455-1473. [DOI: 10.1142/s0192415x20500718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Uric acid nephropathy (UAN) is caused by excessive uric acid, which results in the damage of renal tissue via urate crystals deposition in the kidneys. The roots and rhizomes of Salvia miltiorrhiza Bunge (S. miltiorrhiza) have been clinically used in many prescriptions to treat uric acid-induced renal damage. This study investigates the uricosuric and nephroprotective effects of the ethyl acetate extract of S. miltiorrhiza (EASM) and tanshinone IIA (a major component of S. miltiorrhiza, Tan-IIA) on UAN and explores the underlying molecular mechanism. Both EASM and Tan-IIA significantly decreased serum uric acid (SUA), serum creatinine (SCR), urine uric acid (UUA), and increased urine creatinine (UCR), and blood urea nitrogen (BUN) levels in experimental UAN mice. In adenine and potassium oxonate-induced mice, EASM and Tan-IIA treatment alleviated renal dysfunction and downregulated the expression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS). Moreover, EASM treatment significantly prevented excessive reactive oxygen species (ROS) production in uric acid-induced HK-2 cells and suppressed the expression of nicotinamide adenine dinucleotide phosphate oxidase 4 (NOX4). EASM also suppressed ROS-activated mitogen-activated protein kinases (MAPKs) in vivo and in vitro. These results suggest that both EASM and Tan-IIA demonstrated inhibitory effects on UAN through relieving NOX4-mediated oxidative stress and suppressing MAPK pathways activation.
Collapse
Affiliation(s)
- Xiao-Wei Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Mei Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Lin An
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Ping Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jun Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
168
|
Clinical Application of Melatonin in the Treatment of Cardiovascular Diseases: Current Evidence and New Insights into the Cardioprotective and Cardiotherapeutic Properties. Cardiovasc Drugs Ther 2020; 36:131-155. [PMID: 32926271 DOI: 10.1007/s10557-020-07052-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/04/2020] [Indexed: 12/17/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading global cause of mortality and disability, tending to happen in younger individuals in developed countries. Despite improvements in medical treatments, the therapy and long-term prognosis of CVDs such as myocardial ischemia-reperfusion, atherosclerosis, heart failure, cardiac hypertrophy and remodeling, cardiomyopathy, coronary artery disease, myocardial infarction, and other CVDs threatening human life are not satisfactory enough. Therefore, many researchers are attempting to identify novel potential therapeutic methods for the treatment of CVDs. Melatonin is an anti-inflammatory and antioxidant agent with a wide range of therapeutic properties. Recently, several investigations have been carried out to evaluate its effectiveness and efficiency in CVDs therapy, focusing on mechanistic pathways. Herein, this review aims to summarize current findings of melatonin treatment for CVDs.
Collapse
|
169
|
Roles of Reactive Oxygen Species in Cardiac Differentiation, Reprogramming, and Regenerative Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2102841. [PMID: 32908625 PMCID: PMC7475763 DOI: 10.1155/2020/2102841] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in mechanisms of heart development and regenerative therapies such as the use of pluripotent stem cells. The roles of ROS mediating cell fate are dependent on the intensity of stimuli, cellular context, and metabolic status. ROS mainly act through several targets (such as kinases and transcription factors) and have diverse roles in different stages of cardiac differentiation, proliferation, and maturation. Therefore, further detailed investigation and characterization of redox signaling will help the understanding of the molecular mechanisms of ROS during different cellular processes and enable the design of targeted strategies to foster cardiac regeneration and functional recovery. In this review, we focus on the roles of ROS in cardiac differentiation as well as transdifferentiation (direct reprogramming). The potential mechanisms are discussed in regard to ROS generation pathways and regulation of downstream targets. Further methodological optimization is required for translational research in order to robustly enhance the generation efficiency of cardiac myocytes through metabolic modulations. Additionally, we highlight the deleterious effect of the host's ROS on graft (donor) cells in a paracrine manner during stem cell-based implantation. This knowledge is important for the development of antioxidant strategies to enhance cell survival and engraftment of tissue engineering-based technologies. Thus, proper timing and level of ROS generation after a myocardial injury need to be tailored to ensure the maximal efficacy of regenerative therapies and avoid undesired damage.
Collapse
|
170
|
Cai X, Yang C, Shao L, Zhu H, Wang Y, Huang X, Wang S, Hong L. Targeting NOX 4 by petunidin improves anoxia/reoxygenation-induced myocardium injury. Eur J Pharmacol 2020; 888:173414. [PMID: 32828742 DOI: 10.1016/j.ejphar.2020.173414] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/18/2020] [Accepted: 07/23/2020] [Indexed: 12/30/2022]
Abstract
Oxidative stress is the key factor of myocardial ischemia-reperfusion injury (MIRI). Anthocyanins are considered to be effective anti-oxidants. In this study, we observed the anti-MIRI effect of petunidin, one member of anthocyanins, and further explored its mechanism. In present study, anoxia/reoxygenation (A/R) models were replicated on Langendorff-perfused heart and neonatal rat primary cardiomyocytes by A/R treatment. The hemodynamic parameters of isolated hearts were monitored. The levels of oxidative stress and apoptosis in isolated heart and neonatal rat primary cardiomyocytes were evaluated. The expression levels of NADPH oxidase 2 (NOX 2), NOX 4, B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X (Bax) and cytochrome c were detected by Western Blot. The results showed that petunidin could significantly improve isolated heart function, reduce oxidative stress, inhibit cardiomyocyte apoptosis, up-regulate Bcl-2 protein expression, down-regulate NOX4 and Bax expression, and reduce the level of cytoplasmic cytochrome c after A/R. However, it has no significant effect on NOX 2 protein expression, suggesting that NOX 4 may be the molecular target of petunidin. In vitro, petunidin had shown a consistent effect with that in isolated hearts. It also showed a significant inhibitory effect on reactive oxygen species (ROS) generation. However, the protective effects of petunidin on A/R injury were attenuated by over-expression of NOX 4 in neonatal rat primary cardiomyocytes. These data suggested that the protective effects of petunidin on MIRI may be achieved through targeting NOX 4, thus inhibiting the production of ROS, reducing oxidative stress, and regulating the Bcl-2 pathway to prevent cardiomyocytes apoptosis.
Collapse
Affiliation(s)
- XinYong Cai
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006, PR China
| | - Chunli Yang
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006, PR China
| | - Liang Shao
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006, PR China
| | - HongMin Zhu
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006, PR China
| | - YunXia Wang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006, PR China
| | - Xiao Huang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006, PR China
| | - Shu Wang
- Department of Gerontology, The First Affiliated Hospital of NanChang University, Nanchang, 330006, PR China.
| | - Lang Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006, PR China.
| |
Collapse
|
171
|
Recruitment and maturation of the coronary collateral circulation: Current understanding and perspectives in arteriogenesis. Microvasc Res 2020; 132:104058. [PMID: 32798552 DOI: 10.1016/j.mvr.2020.104058] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/09/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
Abstract
The coronary collateral circulation is a rich anastomotic network of primitive vessels which have the ability to augment in size and function through the process of arteriogenesis. In this review, we evaluate the current understandings of the molecular and cellular mechanisms by which this process occurs, specifically focussing on elevated fluid shear stress (FSS), inflammation, the redox state and gene expression along with the integrative, parallel and simultaneous process by which this occurs. The initiating step of arteriogenesis occurs following occlusion of an epicardial coronary artery, with an increase in FSS detected by mechanoreceptors within the endothelium. This must occur within a 'redox window' where an equilibrium of oxidative and reductive factors are present. These factors initially result in an inflammatory milieu, mediated by neutrophils as well as lymphocytes, with resultant activation of a number of downstream molecular pathways resulting in increased expression of proteins involved in monocyte attraction and adherence; namely vascular cell adhesion molecule 1 (VCAM-1), monocyte chemoattractant protein 1 (MCP-1) and transforming growth factor beta (TGF-β). Once monocytes and other inflammatory cells adhere to the endothelium they enter the extracellular matrix and differentiate into macrophages in an effort to create a favourable environment for vessel growth and development. Activated macrophages secrete inflammatory cytokines such as tumour necrosis factor-α (TNF-α), growth factors such as fibroblast growth factor-2 (FGF-2) and matrix metalloproteinases. Finally, vascular smooth muscle cells proliferate and switch to a contractile phenotype, resulting in an increased diameter and functionality of the collateral vessel, thereby allowing improved perfusion of the distal myocardium subtended by the occluded vessel. This simultaneously reduces FSS within the collateral vessel, inhibiting further vessel growth.
Collapse
|
172
|
Wang CY, Hsiao CY, Tsai KL, Cheng YH. Injectable thermosensitive chitosan-based hydrogel containing ferulic acid for treating peripheral arterial disease. J Tissue Eng Regen Med 2020; 14:1438-1448. [PMID: 32767844 DOI: 10.1002/term.3109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/16/2020] [Accepted: 07/25/2020] [Indexed: 12/20/2022]
Abstract
Peripheral arterial disease (PAD) affects more than 200 million people worldwide. Recent studies suggest that oxidative stress-related inflammation can lead to the initiation and progression of PAD. Ferulic acid (FA) is a natural phenolic compound and has been proven to have antioxidant and angiogenesis effects. In this study, thermosensitive chitosan-gelatin-based hydrogel was used as a delivery vehicle of FA. The effects of hydrogel encapsulating FA (FA-gel) have been demonstrated in vitro and in vivo. The results revealed that the developed hydrogel with porous structure could provide a sustained release of FA. Post-treatment of FA-gel effectively decreased the oxidative stress-induced damage in human umbilical vein endothelial cells via decreasing endogenous reactive oxygen species production, inflammation-related gene expression and apoptosis level. In the mouse hindlimb ischemia model, the results revealed that FA-gel could improve blood flow, muscle regeneration and decreases inflammation in veins. These results suggested that FA-gel may have a therapeutic potential in PAD.
Collapse
Affiliation(s)
- Chien-Ying Wang
- Emergency Department, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Pharmacy & Graduate Institute of Pharmaceutical Technology, Tajen University, Pingtung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | - Kun-Ling Tsai
- Department of Physical Therapy, National Cheng Kung University, Tainan, Taiwan
| | - Yung-Hsin Cheng
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| |
Collapse
|
173
|
Zhang YM, Zhang ZY, Wang RX. Protective Mechanisms of Quercetin Against Myocardial Ischemia Reperfusion Injury. Front Physiol 2020; 11:956. [PMID: 32848878 PMCID: PMC7412593 DOI: 10.3389/fphys.2020.00956] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
Quercetin has attracted more attention in recent years due to its protective role against ischemia/reperfusion injury. Quercetin can alleviate oxidative stress injury through the inhibition of NADPH oxidase and xanthine oxidase, blockage of the Fenton reaction, and scavenging of reactive oxygen species. Quercetin can also exert anti-inflammatory and anti-apoptotic effects by reducing the response to inflammatory factors and inhibiting cell apoptosis. Moreover, it can induce vasodilation effects through the inhibition of endothelin-1 receptors, the enhancement of NO stimulation and the activation of the large-conductance calcium-activated potassium channels. Finally, Quercetin can also antagonize the calcium overload. These multifaceted activities of Quercetin make it a potential therapeutic alternative for the treatment of ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Yu-Min Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Zhen-Ye Zhang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Ru-Xing Wang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
174
|
Meng Y, Du Z, Li Y, Gao P, Song J, Lu Y, Tu P, Jiang Y, Guo X. The synergistic mechanism of total saponins and flavonoids in Notoginseng-Safflower pair against myocardial ischemia uncovered by an integrated metabolomics strategy. Biomed Pharmacother 2020; 130:110574. [PMID: 32739736 DOI: 10.1016/j.biopha.2020.110574] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/11/2020] [Accepted: 07/25/2020] [Indexed: 02/07/2023] Open
Abstract
The Notoginseng-Safflower pair composed of Panax notoginseng (Burk.) F. H. Chen and Carthamus tinctorius L. has remarkable clinical efficacy for preventing and treating cardiovascular diseases in China. Notoginseng total saponins (NS) and Safflower total flavonoids (SF) are the major effective ingredients in Notoginseng and Safflower, respectively. Though our previous study showed that the combination of NS and SF (NS-SF) exhibits significant cardioprotective effects for myocardial ischemia (MI), there might be difference in their action mechanisms. However, the anti-MI characteristics of individual NS and SF remains unclear. Herein, an integrated metabolomics strategy coupled with multiple biological methods were employed to investigate the cardioprotective effects of NS and SF alone or in combination against isoproterenol (ISO)-induced MI and to further explore the synergistic relationship between NS and SF. Our results demonstrated that pretreatments with NS, SF, and NS-SF all showed cardioprotective effects against MI injury and NS-SF exhibited to be the best. Interestingly, the results demonstrated that NS and SF exhibited differentiated metabolic targets and mediators in the glycerophospholipid metabolism. Furthermore, administration of NS alone exhibited greater effects on reversing the elevated the proinflammatory metabolites and mediators in MI rats compared to SF alone. However, individual SF showed greater amelioration of MI-disturbed antioxidant and prooxidative metabolites and better inhibition of the oxidative stress than NS alone. Collectively, our study demonstrated that the capability of NS-SF to regulate both metabolic targets of NS and SF might be the basis of NS-SF to produce a cooperative effect greater than their individual effects that enhance the anti-MI efficacy and provided valuable information for the clinical application of Notoginseng-Safflower pair.
Collapse
Affiliation(s)
- Yuqing Meng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Zhiyong Du
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Yan Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Peng Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Jinyang Song
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Yingyuan Lu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China.
| | - Xiaoyu Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China.
| |
Collapse
|
175
|
Qiu F, Han Y, Shao X, Paulo P, Li W, Zhu M, Tang N, Guo S, Chen Y, Wu H, Zhao D, Liu Y, Chu W. Knockdown of endogenous RNF4 exacerbates ischaemia-induced cardiomyocyte apoptosis in mice. J Cell Mol Med 2020; 24:9545-9559. [PMID: 32722882 PMCID: PMC7520334 DOI: 10.1111/jcmm.15363] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 01/13/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022] Open
Abstract
RNF4, a poly‐SUMO‐specific E3 ubiquitin ligase, is associated with protein degradation, DNA damage repair and tumour progression. However, the effect of RNF4 in cardiomyocytes remains to be explored. Here, we identified the alteration of RNF4 from ischaemic hearts and oxidative stress‐induced apoptotic cardiomyocytes. Upon myocardial infarction (MI) or H2O2/ATO treatment, RNF4 increased rapidly and then decreased gradually. PML SUMOylation and PML nuclear body (PML‐NB) formation first enhanced and then degraded upon oxidative stress. Reactive oxygen species (ROS) inhibitor was able to attenuate the elevation of RNF4 expression and PML SUMOylation. PML overexpression and RNF4 knockdown by small interfering RNA (siRNA) enhanced PML SUMOylation, promoted p53 recruitment and activation and exacerbated H2O2/ATO‐induced cardiomyocyte apoptosis which could be partially reversed by knockdown of p53. In vivo, knockdown of endogenous RNF4 via in vivo adeno‐associated virus infection deteriorated post‐MI structure remodelling including more extensive interstitial fibrosis and severely fractured and disordered structure. Furthermore, knockdown of RNF4 worsened ischaemia‐induced cardiac dysfunction of MI models. Our results reveal a novel myocardial apoptosis regulation model that is composed of RNF4, PML and p53. The modulation of these proteins may provide a new approach to tackling cardiac ischaemia.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Yanna Han
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Xiaoqi Shao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China.,Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, P. R. China
| | - Petro Paulo
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Wenyue Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Mengying Zhu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Nannan Tang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Shuaili Guo
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Yibing Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Han Wu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Dan Zhao
- Departments of Clinical Pharmacy and Cardiology, the 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P.R. China
| | - Yu Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| | - Wenfeng Chu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, P. R. China
| |
Collapse
|
176
|
Caruso G, Fresta CG, Grasso M, Santangelo R, Lazzarino G, Lunte SM, Caraci F. Inflammation as the Common Biological Link Between Depression and Cardiovascular Diseases: Can Carnosine Exert a Protective Role? Curr Med Chem 2020; 27:1782-1800. [PMID: 31296155 DOI: 10.2174/0929867326666190712091515] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/15/2019] [Accepted: 02/05/2019] [Indexed: 01/03/2023]
Abstract
Several epidemiological studies have clearly shown the high co-morbidity between depression and Cardiovascular Diseases (CVD). Different studies have been conducted to identify the common pathophysiological events of these diseases such as the overactivation of the hypothalamic- pituitary-adrenal axis and, most importantly, the dysregulation of immune system which causes a chronic pro-inflammatory status. The biological link between depression, inflammation, and CVD can be related to high levels of pro-inflammatory cytokines, such as IL-1β, TNF-α, and IL-6, released by macrophages which play a central role in the pathophysiology of both depression and CVD. Pro-inflammatory cytokines interfere with many of the pathophysiological mechanisms relevant to depression by upregulating the rate-limiting enzymes in the metabolic pathway of tryptophan and altering serotonin metabolism. These cytokines also increase the risk to develop CVD, because activation of macrophages under this pro-inflammatory status is closely associated with endothelial dysfunction and oxidative stress, a preamble to atherosclerosis and atherothrombosis. Carnosine (β-alanyl-L-histidine) is an endogenous dipeptide which exerts a strong antiinflammatory activity on macrophages by suppressing reactive species and pro-inflammatory cytokines production and altering pro-inflammatory/anti-inflammatory macrophage polarization. This dipeptide exhibits antioxidant properties scavenging reactive species and preventing oxidative stress-induced pathologies such as CVD. In the present review we will discuss the role of oxidative stress and chronic inflammation as common pathophysiological events both in depression and CVD and the preclinical and clinical evidence on the protective effect of carnosine in both diseases as well as the therapeutic potential of this dipeptide in depressed patients with a high co-morbidity of cardiovascular diseases.
Collapse
Affiliation(s)
- Giuseppe Caruso
- Oasi Research Institute - IRCCS, Via Conte Ruggero, 73, Troina 94018, Italy
| | - Claudia G Fresta
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania 95125, Italy
| | - Margherita Grasso
- Oasi Research Institute - IRCCS, Via Conte Ruggero, 73, Troina 94018, Italy.,Department of Drug Sciences, University of Catania, Catania 95125, Italy
| | - Rosa Santangelo
- Department of Drug Sciences, University of Catania, Catania 95125, Italy
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Catania 95125, Italy
| | - Susan M Lunte
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence 66045, Kansas, United States.,Department of Pharmaceutical Chemistry, University of Kansas, Lawrence 66045, Kansas, United States.,Department of Chemistry, University of Kansas, Lawrence 66045, Kansas, United States
| | - Filippo Caraci
- Oasi Research Institute - IRCCS, Via Conte Ruggero, 73, Troina 94018, Italy.,Department of Drug Sciences, University of Catania, Catania 95125, Italy
| |
Collapse
|
177
|
Perrone MA, Babu Dasari J, Intorcia A, Gualtieri P, Marche M, Di Luozzo M, Merra G, Bernardini S, Romeo F, Sergi D. Phenotypic classification and biochemical profile of obesity for cardiovascular prevention. GAZZETTA MEDICA ITALIANA ARCHIVIO PER LE SCIENZE MEDICHE 2020. [DOI: 10.23736/s0393-3660.20.04259-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
178
|
Kapil V, Khambata RS, Jones DA, Rathod K, Primus C, Massimo G, Fukuto JM, Ahluwalia A. The Noncanonical Pathway for In Vivo Nitric Oxide Generation: The Nitrate-Nitrite-Nitric Oxide Pathway. Pharmacol Rev 2020; 72:692-766. [PMID: 32576603 DOI: 10.1124/pr.120.019240] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
In contrast to nitric oxide, which has well established and important roles in the regulation of blood flow and thrombosis, neurotransmission, the normal functioning of the genitourinary system, and the inflammation response and host defense, its oxidized metabolites nitrite and nitrate have, until recently, been considered to be relatively inactive. However, this view has been radically revised over the past decade and more. Much evidence has now accumulated demonstrating that nitrite serves as a storage form of nitric oxide, releasing nitric oxide preferentially under acidic and/or hypoxic conditions but also occurring under physiologic conditions: a phenomenon that is catalyzed by a number of distinct mammalian nitrite reductases. Importantly, preclinical studies demonstrate that reduction of nitrite to nitric oxide results in a number of beneficial effects, including vasodilatation of blood vessels and lowering of blood pressure, as well as cytoprotective effects that limit the extent of damage caused by an ischemia/reperfusion insult, with this latter issue having been translated more recently to the clinical setting. In addition, research has demonstrated that the other main metabolite of the oxidation of nitric oxide (i.e., nitrate) can also be sequentially reduced through processing in vivo to nitrite and then nitrite to nitric oxide to exert a range of beneficial effects-most notably lowering of blood pressure, a phenomenon that has also been confirmed recently to be an effective method for blood pressure lowering in patients with hypertension. This review will provide a detailed description of the pathways involved in the bioactivation of both nitrate and nitrite in vivo, their functional effects in preclinical models, and their mechanisms of action, as well as a discussion of translational exploration of this pathway in diverse disease states characterized by deficiencies in bioavailable nitric oxide. SIGNIFICANCE STATEMENT: The past 15 years has seen a major revision in our understanding of the pathways for nitric oxide synthesis in the body with the discovery of the noncanonical pathway for nitric oxide generation known as the nitrate-nitrite-nitric oxide pathway. This review describes the molecular components of this pathway, its role in physiology, potential therapeutics of targeting this pathway, and their impact in experimental models, as well as the clinical translation (past and future) and potential side effects.
Collapse
Affiliation(s)
- V Kapil
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - R S Khambata
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - D A Jones
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - K Rathod
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - C Primus
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - G Massimo
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - J M Fukuto
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| | - A Ahluwalia
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom (V.K., R.S.K., D.A.J., K.R., C.P., G.M., A.A.) and Department of Chemistry, Sonoma State University, Rohnert Park, California (J.M.F.)
| |
Collapse
|
179
|
Jeon SY, Kim MR, Yu SH, Kim MJ, Shim KS, Shin E, Lee JJ, Lee YC. Combined Extract of Vitis vinifera L. and Centella asiatica Synergistically Attenuates Oxidative Damage Induced by Hydrogen Peroxide in Human Umbilical Vein Endothelial Cells. Prev Nutr Food Sci 2020; 25:173-183. [PMID: 32676469 PMCID: PMC7333004 DOI: 10.3746/pnf.2020.25.2.173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/20/2020] [Indexed: 11/06/2022] Open
Abstract
Endothelial cell injury caused by oxidative stress is a critical factor in the initial stage of vascular diseases. Thus, identification of more effective antioxidants is a promising strategy to protect against endothelial cell injury. Recently, synergistic effects between phytochemicals have received renewed attention for their role in the treatment of various diseases. Vitis vinifera L. and Centella asiatica are well-known medicinal plants with various biological effects. However, the combination of the two has not previously been studied. Here, we investigated the effects of V. vinifera L. leaf and C. asiatica extract combination (VCEC), a standardized herbal blend comprising V. vinifera L. leaf extract (VE) and C. asiatica extract (CE), for its antioxidant activity and for the protection of endothelial cells against hydrogen peroxide (H2O2)-mediated oxidative damage in human umbilical vein endothelial cells (HUVECs). VCEC showed higher antioxidant activity than VE or CE in oxygen radical antioxidant capacity assays. In HUVECs, VCEC significantly suppressed increases in the production of intracellular reactive oxygen species, decreased levels of nitric oxide and vascular endothelial-cadherin, and increased endothelial hyperpermeability triggered by H2O2. Treatment with VE or CE alone ameliorated HUVEC injury in a pattern similar to VCEC, although their effects were significantly weaker than VCEC. Overall, VCEC exhibited a substantial synergistic effect on protecting endothelial cells against oxidative damage through its antioxidant activity. Therefore, VCEC could be developed as a potential agent for reducing the risk of vascular diseases related to oxidative stress.
Collapse
Affiliation(s)
- Se Yeong Jeon
- R&D Center, Naturetech Co., Ltd., Chungnam 31257, Korea
| | - Mi Ran Kim
- R&D Center, Naturetech Co., Ltd., Chungnam 31257, Korea
| | - Su Hyun Yu
- R&D Center, Naturetech Co., Ltd., Chungnam 31257, Korea
| | | | | | | | - Jeong Jun Lee
- R&D Center, Naturetech Co., Ltd., Chungnam 31257, Korea
| | | |
Collapse
|
180
|
Jeon SY, Kim MR, Lee EO, Jeon BH, Lee JJ, Lee YC. Effect of a new herbal composition comprised of red clover and hop extract on human endothelial cell damage and vasorelaxant activity. J Food Biochem 2020; 44:e13314. [PMID: 32542699 DOI: 10.1111/jfbc.13314] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/24/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022]
Abstract
Hormone replacement therapy may cause various side effects, including enhancing the risk of cardiovascular disease (CVD) in postmenopausal women. Here, we investigated the effect of red clover and hop extract combination (RHEC) on estrogen receptor (ER) binding and endothelial function of human umbilical vein endothelial cells (HUVECs) to develop an herbal agent for reducing the risk of CVDs. In ER competitor assay, RHEC showed binding affinity toward ERα and ERβ with IC50 values of 5.92 µg/ml and 1.66 µg/ml, respectively. In HUVECs, RHEC significantly increased the cell viability and reduced the reactive oxygen species production against oxidative stress-induced damage. We also showed that RHEC increased the NO production through upregulating the endothelial nitric oxide synthase expression via ER activation in estrogen depleted condition. In particular, RHEC showed greater efficacy with increase in NO and decrease in endothelin-1 than red clover or hop treatment alone. Additionally, 0.3-0.5 mg/ml of RHEC-induced vasorelaxation of rat aortic rings precontracted by phenylephrine. PRACTICAL APPLICATIONS: Recently, a large interest has grown in the synergistic effects of phytochemicals for better therapies to treat various diseases. Red clover and hop are well-known edible plants which are widely used to help relieve postmenopausal symptoms including CVD. However, their combination has not been studied so far. For the first time, we demonstrated that RHEC, a new herbal combination comprising the extracts from red clover and hop, appeared to be effective in protection of endothelial function against oxidative stress and estrogen depletion. Therefore, RHEC could be a potent herbal agent for reducing the risk of endothelial damage.
Collapse
Affiliation(s)
- Se Yeong Jeon
- R&D Center, Naturetech Co., Ltd, 450-86, Maebong-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, Republic of Korea
| | - Mi Ran Kim
- R&D Center, Naturetech Co., Ltd, 450-86, Maebong-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, Republic of Korea
| | - Eun Ok Lee
- Department of Physiology, Infection Signaling Network Research Center, Research Institute for Medical Sciences, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Byeong Hwa Jeon
- Department of Physiology, Infection Signaling Network Research Center, Research Institute for Medical Sciences, School of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong Jun Lee
- R&D Center, Naturetech Co., Ltd, 450-86, Maebong-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, Republic of Korea
| | - Young Chul Lee
- R&D Center, Naturetech Co., Ltd, 450-86, Maebong-Ro, Dongnam-Gu, Cheonan-Si, Chungnam, Republic of Korea
| |
Collapse
|
181
|
Waldman M, Arad M, Abraham NG, Hochhauser E. The Peroxisome Proliferator-Activated Receptor-Gamma Coactivator-1α-Heme Oxygenase 1 Axis, a Powerful Antioxidative Pathway with Potential to Attenuate Diabetic Cardiomyopathy. Antioxid Redox Signal 2020; 32:1273-1290. [PMID: 32027164 PMCID: PMC7232636 DOI: 10.1089/ars.2019.7989] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023]
Abstract
Significance: From studies of diabetic animal models, the downregulation of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α)-heme oxygenase 1 (HO-1) axis appears to be a crucial event in the development of obesity and diabetic cardiomyopathy (DCM). In this review, we discuss the role of metabolic and biochemical stressors in the rodent and human pathophysiology of DCM. A crucial contributor for many cardiac pathologies is excessive production of reactive oxygen species (ROS) pathologies, which lead to extensive cellular damage by impairing mitochondrial function and directly oxidizing DNA, proteins, and lipid membranes. We discuss the role of ROS production and inflammatory pathways with multiple contributing and confounding factors leading to DCM. Recent Advances: The relevant biochemical pathways that are critical to a therapeutic approach to treat DCM, specifically caloric restriction and its relation to the PGC-1α-HO-1 axis in the attenuation of DCM, are elucidated. Critical Issues: The increased prevalence of diabetes mellitus type 2, a major contributor to unique cardiomyopathy characterized by cardiomyocyte hypertrophy with no effective clinical treatment. This review highlights the role of mitochondrial dysfunction in the development of DCM and potential oxidative targets to attenuate oxidative stress and attenuate DCM. Future Directions: Targeting the PGC-1α-HO-1 axis is a promising approach to ameliorate DCM through improvement in mitochondrial function and antioxidant defenses. A pharmacological inducer to activate PGC-1α and HO-1 described in this review may be a promising therapeutic approach in the clinical setting.
Collapse
Affiliation(s)
- Maayan Waldman
- Cardiac Research Laboratory, Felsenstein Medical Research Institute at Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
- Cardiac Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Ramat Gan, Israel
| | - Michael Arad
- Cardiac Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Ramat Gan, Israel
| | - Nader G. Abraham
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Edith Hochhauser
- Cardiac Research Laboratory, Felsenstein Medical Research Institute at Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
182
|
Ozaki Tan SJ, Floriano JF, Nicastro L, Emanueli C, Catapano F. Novel Applications of Mesenchymal Stem Cell-derived Exosomes for Myocardial Infarction Therapeutics. Biomolecules 2020; 10:E707. [PMID: 32370160 PMCID: PMC7277090 DOI: 10.3390/biom10050707] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality and morbidity globally, representing approximately a third of all deaths every year. The greater part of these cases is represented by myocardial infarction (MI), or heart attack as it is better known, which occurs when declining blood flow to the heart causes injury to cardiac tissue. Mesenchymal stem cells (MSCs) are multipotent stem cells that represent a promising vector for cell therapies that aim to treat MI due to their potent regenerative effects. However, it remains unclear the extent to which MSC-based therapies are able to induce regeneration in the heart and even less clear the degree to which clinical outcomes could be improved. Exosomes, which are small extracellular vesicles (EVs) known to have implications in intracellular communication, derived from MSCs (MSC-Exos), have recently emerged as a novel cell-free vector that is capable of conferring cardio-protection and regeneration in target cardiac cells. In this review, we assess the current state of research of MSC-Exos in the context of MI. In particular, we place emphasis on the mechanisms of action by which MSC-Exos accomplish their therapeutic effects, along with commentary on the current difficulties faced with exosome research and the ongoing clinical applications of stem-cell derived exosomes in different medical contexts.
Collapse
Affiliation(s)
- Sho Joseph Ozaki Tan
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| | - Juliana Ferreria Floriano
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
- Botucatu Medical School, Sao Paulo State University, Botucatu 18618687, Brazil
| | - Laura Nicastro
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| | - Francesco Catapano
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| |
Collapse
|
183
|
Kračun D, Klop M, Knirsch A, Petry A, Kanchev I, Chalupsky K, Wolf CM, Görlach A. NADPH oxidases and HIF1 promote cardiac dysfunction and pulmonary hypertension in response to glucocorticoid excess. Redox Biol 2020; 34:101536. [PMID: 32413743 PMCID: PMC7226895 DOI: 10.1016/j.redox.2020.101536] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/31/2020] [Accepted: 04/04/2020] [Indexed: 12/22/2022] Open
Abstract
Cardiovascular side effects are frequent problems accompanying systemic glucocorticoid therapy, although the underlying mechanisms are not fully resolved. Reactive oxygen species (ROS) have been shown to promote various cardiovascular diseases although the link between glucocorticoid and ROS signaling has been controversial. As the family of NADPH oxidases has been identified as important source of ROS in the cardiovascular system we investigated the role of NADPH oxidases in response to the synthetic glucocorticoid dexamethasone in the cardiovascular system in vitro and in vivo in mice lacking functional NADPH oxidases due to a mutation in the gene coding for the essential NADPH oxidase subunit p22phox. We show that dexamethasone induced NADPH oxidase-dependent ROS generation, leading to vascular proliferation and angiogenesis due to activation of the transcription factor hypoxia-inducible factor-1 (HIF1). Chronic treatment of mice with low doses of dexamethasone resulted in the development of systemic hypertension, cardiac hypertrophy and left ventricular dysfunction, as well as in pulmonary hypertension and pulmonary vascular remodeling. In contrast, mice deficient in p22phox-dependent NADPH oxidases were protected against these cardiovascular side effects. Mechanistically, dexamethasone failed to upregulate HIF1α levels in these mice, while vascular HIF1α deficiency prevented pulmonary vascular remodeling. Thus, p22phox-dependent NADPH oxidases and activation of the HIF pathway are critical elements in dexamethasone-induced cardiovascular pathologies and might provide interesting targets to limit cardiovascular side effects in patients on chronic glucocorticoid therapy.
Collapse
Affiliation(s)
- Damir Kračun
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Mathieu Klop
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Anna Knirsch
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Andreas Petry
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Ivan Kanchev
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany
| | - Karel Chalupsky
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany; Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Cordula M Wolf
- Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, Department of Pediatric Cardiology and Congenital Heart Diseases, German Heart Center Munich at the Technical University Munich, Munich, 80636, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
184
|
Soni H, Yakimkova T, Matthews AT, Amartey PK, Read RW, Buddington RK, Adebiyi A. Early onset of renal oxidative stress in small for gestational age newborn pigs. Redox Rep 2020; 24:10-16. [PMID: 30907266 PMCID: PMC6448771 DOI: 10.1080/13510002.2019.1596429] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Oxidative stress, a common feature in cardiovascular and renal disease is associated with the causes and consequences of fetal growth restriction. Hence, renal redox status is likely an early determinant of morbidity in small-for-gestational-age (SGA) infants. In this study, we examined renal oxidative stress in naturally-farrowed SGA newborn pigs. METHODS We studied SGA newborn pigs with 52% less body weight and 59% higher brain/liver weight ratio compared with their appropriate-for-gestational-age (AGA) counterparts. RESULTS The kidneys of the SGA newborn pigs weighed 56% less than the AGA group. The glomerular cross-sectional area was also smaller in the SGA group. SGA newborn pigs exhibited increased renal lipid peroxidation, reduced kidney and urine total antioxidant capacity, and increased renal nitrotyrosine immunostaining. Whereas the protein expression level of NADPH oxidase (NOX)2 was unchanged, NOX4 expression was significantly higher in SGA kidneys. The level of serum potassium was lower, but serum sodium and creatinine were similar in SGA compared with AGA newborn pigs. The serum concentrations of C-reactive protein and NGAL, the biomarkers of inflammation and early acute kidney injury were significantly elevated in the SGA group. CONCLUSION Early induction of oxidative stress may contribute to the onset of kidney injury in growth-restricted infants.
Collapse
Affiliation(s)
- Hitesh Soni
- a Department of Physiology , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Taisiya Yakimkova
- b School of Health Studies , University of Memphis , Memphis , TN , USA
| | - Anberitha T Matthews
- a Department of Physiology , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Paul K Amartey
- a Department of Physiology , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Robert W Read
- c Department of Biological Sciences , University of Memphis , Memphis , TN , USA
| | - Randal K Buddington
- a Department of Physiology , University of Tennessee Health Science Center , Memphis , TN , USA.,b School of Health Studies , University of Memphis , Memphis , TN , USA.,d College of Nursing , University of Tennessee Health Science Center , Memphis , TN , USA
| | - Adebowale Adebiyi
- a Department of Physiology , University of Tennessee Health Science Center , Memphis , TN , USA
| |
Collapse
|
185
|
Ping Z, Peng Y, Lang H, Xinyong C, Zhiyi Z, Xiaocheng W, Hong Z, Liang S. Oxidative Stress in Radiation-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3579143. [PMID: 32190171 PMCID: PMC7071808 DOI: 10.1155/2020/3579143] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/03/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023]
Abstract
There is a distinct increase in the risk of heart disease in people exposed to ionizing radiation (IR). Radiation-induced heart disease (RIHD) is one of the adverse side effects when people are exposed to ionizing radiation. IR may come from various forms, such as diagnostic imaging, radiotherapy for cancer treatment, nuclear disasters, and accidents. However, RIHD was mainly observed after radiotherapy for chest malignant tumors, especially left breast cancer. Radiation therapy (RT) has become one of the main ways to treat all kinds of cancer, which is used to reduce the recurrence of cancer and improve the survival rate of patients. The potential cause of radiation-induced cardiotoxicity is unclear, but it may be relevant to oxidative stress. Oxidative stress, an accumulation of reactive oxygen species (ROS), disrupts intracellular homeostasis through chemical modification and damages proteins, lipids, and DNA; therefore, it results in a series of related pathophysiological changes. The purpose of this review was to summarise the studies of oxidative stress in radiotherapy-induced cardiotoxicity and provide prevention and treatment methods to reduce cardiac damage.
Collapse
Affiliation(s)
- Zhang Ping
- Department of Neurology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Yang Peng
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Hong Lang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Cai Xinyong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Zeng Zhiyi
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Wu Xiaocheng
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Zeng Hong
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| | - Shao Liang
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, 330006 Jiangxi, China
| |
Collapse
|
186
|
Flahault A, Paquette K, Fernandes RO, Delfrate J, Cloutier A, Henderson M, Lavoie JC, Mâsse B, Nuyt AM, Luu TM, Alos N, Bertagnolli M, Bigras JL, Curnier D, Dartora DR, Ducruet T, El-Jalbout R, Girard-Bock C, Gyger G, Hamel P, Lapeyraque AL, Mian MOR, Orlando V, Xie LF. Increased Incidence but Lack of Association Between Cardiovascular Risk Factors in Adults Born Preterm. Hypertension 2020; 75:796-805. [DOI: 10.1161/hypertensionaha.119.14335] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Preterm birth incurs an increased risk of early cardiovascular events and death. In the general population, cardiovascular risk factors cluster in the context of inflammation and oxidative stress. Whether this also occurs in young adults born preterm is unknown. We analyzed 101 healthy young adults (ages 18–29) born preterm (≤29 weeks of gestation) and 105 full-term controls, predominantly (90%) white. They underwent a comprehensive clinical and biological evaluation, including measurement of blood pressure, lung function (spirometry), glucose metabolism (fasting glucose, glycated hemoglobin, and oral glucose tolerance test), as well as biomarkers of inflammation and oxidative stress. Individuals born preterm were at higher risk than those born full-term of stage ≥1 hypertension (adjusted odds ratio, 2.91 [95% CI, 1.51–5.75]), glucose intolerance (adjusted odds ratio, 2.22 [95% CI, 1.13–4.48]), and airflow limitation (adjusted odds ratio, 3.47 [95% CI, 1.76–7.12]). Hypertension was strongly associated with adiposity and with glucose intolerance in participants born full-term but not in those born preterm. We did not find any group difference in levels of biomarkers of inflammation and oxidative stress. In individuals born preterm, inflammation, and oxidative stress were not related to hypertension or glucose intolerance but were associated with adiposity. In those born preterm, cardiovascular risk factors were not related to each other suggesting different pathophysiological pathways leading to the development of cardiovascular risk following preterm birth. Clinicians should consider screening for these abnormalities irrespectively of other risk factors in this at-risk population.
Clinical Trial Registration
URL:
http://www.clinicaltrials.gov
. Unique identifier: NCT03261609.
Collapse
Affiliation(s)
- Adrien Flahault
- From the Department of Pediatrics (A.F., K.P., R.O.F., J.D., A.C., M.H., A.M.N., T.M.L.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | - Katryn Paquette
- From the Department of Pediatrics (A.F., K.P., R.O.F., J.D., A.C., M.H., A.M.N., T.M.L.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | - Rafael Oliveira Fernandes
- From the Department of Pediatrics (A.F., K.P., R.O.F., J.D., A.C., M.H., A.M.N., T.M.L.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | - Jacques Delfrate
- From the Department of Pediatrics (A.F., K.P., R.O.F., J.D., A.C., M.H., A.M.N., T.M.L.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | - Anik Cloutier
- From the Department of Pediatrics (A.F., K.P., R.O.F., J.D., A.C., M.H., A.M.N., T.M.L.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | - Mélanie Henderson
- From the Department of Pediatrics (A.F., K.P., R.O.F., J.D., A.C., M.H., A.M.N., T.M.L.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | - Jean-Claude Lavoie
- Department of Nutrition (J.-C.L.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | - Benoît Mâsse
- Department of Social and Preventive Medicine, School of Public Health (B.M.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | - Anne Monique Nuyt
- From the Department of Pediatrics (A.F., K.P., R.O.F., J.D., A.C., M.H., A.M.N., T.M.L.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | - Thuy Mai Luu
- From the Department of Pediatrics (A.F., K.P., R.O.F., J.D., A.C., M.H., A.M.N., T.M.L.), Sainte-Justine University Hospital and Research Center, Université de Montréal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Signorelli SS, Vanella L, Abraham NG, Scuto S, Marino E, Rocic P. Pathophysiology of chronic peripheral ischemia: new perspectives. Ther Adv Chronic Dis 2020; 11:2040622319894466. [PMID: 32076496 PMCID: PMC7003198 DOI: 10.1177/2040622319894466] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022] Open
Abstract
Peripheral arterial disease (PAD) affects individuals particularly over 65 years old in the more advanced countries. Hemodynamic, inflammatory, and oxidative mechanisms interact in the pathophysiological scenario of this chronic arterial disease. We discuss the hemodynamic, muscle tissue, and oxidative stress (OxS) conditions related to chronic ischemia of the peripheral arteries. This review summarizes the results of evaluating both metabolic and oxidative markers, and also therapy to counteract OxS. In conclusion, we believe different pathways should be highlighted to discover new drugs to treat patients suffering from PAD.
Collapse
Affiliation(s)
- Salvatore Santo Signorelli
- Department of Clinical and Experimental Medicine, University of Catania, University Hospital ‘G. Rodolico’, Catania, 95124, Italy
| | - Luca Vanella
- Department of Drug Science, University of Catania, Catania, Italy
| | - Nader G. Abraham
- Departments of Medicine, Pharmacology and Gastroenterology, New York Medical College, Valhalla, NY, USA
| | - Salvatore Scuto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Elisa Marino
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Petra Rocic
- Departments of Medicine, Pharmacology and Gastroenterology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
188
|
Miao J, Xu M, Kuang Y, Pan S, Hou J, Cao P, Duan X, Chang Y, Hasem H, Zhou N, Tan K, Fan Y. Deferasirox protects against hydrogen peroxide-induced cell apoptosis by inhibiting ubiquitination and degradation of p21 WAF1/CIP1. Biochem Biophys Res Commun 2020; 524:736-743. [PMID: 32035614 DOI: 10.1016/j.bbrc.2020.01.155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 12/29/2022]
Abstract
Deferasirox (DFX) is an iron chelator approved for the treatment of iron overload diseases. However, the role of DFX in oxidative stress-induced cell apoptosis and the exact molecular mechanisms underlying these processes remain poorly understood and require further investigation. In this study, we found that DFX rendered resistant to H2O2-induced apoptosis in HEK293T cells, reduced the intracellular levels of the labile iron pool (LIP) and oxidative stress induced by H2O2. Furthermore, DFX inhibited the ubiquitination and degradation of the cyclin-dependent kinase inhibitor p21WAF1/CIP1 (p21) via modulation of the interaction of p21 with SCF-Skp2. DFX also showed the inhibition effect on the activation of c-Jun N-terminal kinase (JNK), pro-caspase-3 and related mitochondrial apoptosis pathway induced by H2O2. These results provide novel insights into the molecular mechanism underpinning iron-mediated oxidative stress and apoptosis, and they may represent a promising target for therapeutic interventions in related pathological conditions.
Collapse
Affiliation(s)
- Junhua Miao
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China
| | - Mutao Xu
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China
| | - Yuhuan Kuang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China
| | - Shuhong Pan
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China
| | - Jianyuan Hou
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China
| | - Pengxiu Cao
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China
| | - Xianglin Duan
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China
| | - Yanzhong Chang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China
| | - Habelhah Hasem
- Department of Pathology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, United States
| | - Nan Zhou
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China; Department of Gynecolog, Xingtai People's Hospital, Xingtai, 054031, PR China
| | - Ke Tan
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China.
| | - Yumei Fan
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Key Laboratory of Molecular and Cellular Biology of Ministry of Education, College of Life Science, Hebei Normal University, Shijiazhuang, 050024, PR China; Department of Pathology, Carver College of Medicine, The University of Iowa, Iowa City, IA, 52242, United States.
| |
Collapse
|
189
|
A Novel Pectic Polysaccharide of Jujube Pomace: Structural Analysis and Intracellular Antioxidant Activities. Antioxidants (Basel) 2020; 9:antiox9020127. [PMID: 32024245 PMCID: PMC7070808 DOI: 10.3390/antiox9020127] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/24/2020] [Accepted: 01/30/2020] [Indexed: 01/09/2023] Open
Abstract
After extraction from jujube pomace and purification by two columns (DEAE-Sepharose Fast Flow and Sepharcyl S-300), the structure of SAZMP4 was investigated by HPGPC, GC, FI-IR, GC-MS, NMR, SEM, and AFM. Analysis determined that SAZMP4 (Mw = 28.94 kDa) was a pectic polysaccharide mainly containing 1,4-linked GalA (93.48%) with side chains of 1,2,4-linked Rha and 1,3,5-linked Ara and terminals of 1-linked Rha and 1-linked Ara, which might be the homogalacturonan (HG) type with side chains of the RG-I type, corresponding to the results of NMR. In AFM and SEM images, self-assembly and aggregation of SAZMP4 were respectively observed indicating its structural features. The antioxidant activity of SAZMP4 against H2O2-induced oxidative stress in Caco-2 cells was determined by activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) as well as malondialdehyde (MDA) and reactive oxygen species (ROS) levels, indicating SAZMP4 can be a natural antioxidant. Also, a better water retention capacity and thermal stability of SAZMP4 was observed based on DSC analysis, which could be applied in food industry as an additive.
Collapse
|
190
|
Parsanathan R, Jain SK. Novel Invasive and Noninvasive Cardiac-Specific Biomarkers in Obesity and Cardiovascular Diseases. Metab Syndr Relat Disord 2020; 18:10-30. [PMID: 31618136 PMCID: PMC7041332 DOI: 10.1089/met.2019.0073] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of fatality and disability worldwide regardless of gender. Obesity has reached epidemic proportions in population across different regions. According to epidemiological studies, CVD risk markers in childhood obesity are one of the significant risk factors for adulthood CVD, but have received disproportionally little attention. This review has examined the evidence for the presence of traditional cardiac biomarkers (nonspecific; lactate dehydrogenase, alanine aminotransferase, aspartate aminotransferase, creatine kinase, myoglobulin, glycogen phosphorylase isoenzyme BB, myosin light chains, ST2, and ischemia-modified albumin) and novel emerging cardiac-specific biomarkers (cardiac troponins, natriuretic peptides, heart-type fatty acid-binding protein, and miRNAs). Besides, noninvasive anatomical and electrophysiological markers (carotid intima-media thickness, coronary artery calcification, and heart rate variability) in CVDs and obesity are also discussed. Modifiable and nonmodifiable risk factors associated with metabolic syndrome in the progression of CVD, such as obesity, diabetes, hypertension, dyslipidemia, oxidative stress, inflammation, and adipocytokines are also outlined. These underlying prognostic risk factors predict the onset of future microvascular and macrovascular complications. The understanding of invasive and noninvasive cardiac-specific biomarkers and the risk factors may yield valuable insights into the pathophysiology and prevention of CVD in a high-risk obese population at an early stage.
Collapse
Affiliation(s)
- Rajesh Parsanathan
- Department of Pediatrics and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Sushil K. Jain
- Department of Pediatrics and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| |
Collapse
|
191
|
Shukla H, Chitrakar R, Bibi HA, Gaje G, Koucheki A, Trush MA, Zhu H, Li YR, Jia Z. Reactive oxygen species production by BP-1,6-quinone and its effects on the endothelial dysfunction: Involvement of the mitochondria. Toxicol Lett 2020; 322:120-130. [PMID: 31953210 DOI: 10.1016/j.toxlet.2020.01.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 01/11/2023]
Abstract
Strong epidemiological evidence supports the association between increased air pollution and the risk of developing atherosclerotic cardiovascular diseases (CVDs). However, the mechanism remains unclear. As an environmental air pollutant and benzo-a-pyrene (BP) metabolite, BP-1,6-quinone (BP-1,6-Q) is present in the particulate phase of air pollution. This study was undertaken to examine the redox activity of BP-1,6-Q and mechanisms associated with it using EA.hy926 endothelial cells. BP-1,6-Q at 0.01-1 μM significantly stimulated the production of reactive oxygen species (ROS)·in intact cells and isolated mitochondria. Furthermore, BP-1,6-Q-induced ROS was altered by mitochondrial electron transport chain (METC) inhibitors of complex I (rotenone) and complex III (antimycin A), denoting the involvement of mitochondrial electron transport chain (METC) in BP-1,6-Q mediated ROS production. In METC deficient cells, interestingly, BP-1,6-Q-mediated ROS production was enhanced, suggesting that overproduction of ROS by BP-1,6-Q is not only produced from mitochondria but can also be from the cell outside of mitochondria (extramitochondrial). BP-1,6-Q also triggered endothelial-monocyte interaction and stimulated expression of vascular adhesion molecule-1 (VCAM-1). In conclusion, these results demonstrate that BP-1,6-Q can generate ROS within both mitochondria and outside of mitochondria, resulting in stimulation of adhesion of monocytes to endothelial cells, a key event in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Halley Shukla
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Rojin Chitrakar
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Humaira A Bibi
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Gabriella Gaje
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Ashkon Koucheki
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Michael A Trush
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Hong Zhu
- Campbell University School of Osteopathic Medicine, Buies Creek, NC, United States
| | - Y Robert Li
- Department of Biology, University of North Carolina at Greensboro, NC, United States; Campbell University School of Osteopathic Medicine, Buies Creek, NC, United States.
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, NC, United States.
| |
Collapse
|
192
|
Oxidative Modifications in Advanced Atherosclerotic Plaques: A Focus on In Situ Protein Sulfhydryl Group Oxidation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6169825. [PMID: 31998439 PMCID: PMC6973184 DOI: 10.1155/2020/6169825] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 12/21/2022]
Abstract
Although oxidative stress has been long associated with the genesis and progression of the atherosclerotic plaque, scanty data on its in situ effects on protein sulfhydryl group modifications are available. Within the arterial wall, protein sulfhydryls and low-molecular-weight (LMW) thiols are involved in the cell regulation of both Reactive Oxygen Species (ROS) and Reactive Nitrogen Species (RNS) levels and are a target for several posttranslational oxidative modifications that take place inside the atherosclerotic plaque, probably contributing to both atherogenesis and atherosclerotic plaque progression towards complicated lesions. Advanced carotid plaques are characterized by very high intraplaque GSH levels, due to cell lysis during apoptotic and/or necrotic events, probably responsible for the altered equilibrium among protein sulfhydryls and LMW thiols. Some lines of evidence show that the prooxidant environment present in atherosclerotic tissue could modify filtered proteins also by protein-SH group oxidation, and demonstrate that particularly albumin, once filtered, represents a harmful source of homocysteine and cysteinylglycine inside the plaque. The oxidative modification of protein sulfhydryls, with particular emphasis to protein thiolation by LMW thiols and its association with atherosclerosis, is the main topic of this review.
Collapse
|
193
|
Kura B, Szeiffova Bacova B, Kalocayova B, Sykora M, Slezak J. Oxidative Stress-Responsive MicroRNAs in Heart Injury. Int J Mol Sci 2020; 21:E358. [PMID: 31948131 PMCID: PMC6981696 DOI: 10.3390/ijms21010358] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are important molecules in the living organisms as a part of many signaling pathways. However, if overproduced, they also play a significant role in the development of cardiovascular diseases, such as arrhythmia, cardiomyopathy, ischemia/reperfusion injury (e.g., myocardial infarction and heart transplantation), and heart failure. As a result of oxidative stress action, apoptosis, hypertrophy, and fibrosis may occur. MicroRNAs (miRNAs) represent important endogenous nucleotides that regulate many biological processes, including those involved in heart damage caused by oxidative stress. Oxidative stress can alter the expression level of many miRNAs. These changes in miRNA expression occur mainly via modulation of nuclear factor erythroid 2-related factor 2 (Nrf2), sirtuins, calcineurin/nuclear factor of activated T cell (NFAT), or nuclear factor kappa B (NF-κB) pathways. Up until now, several circulating miRNAs have been reported to be potential biomarkers of ROS-related cardiac diseases, including myocardial infarction, hypertrophy, ischemia/reperfusion, and heart failure, such as miRNA-499, miRNA-199, miRNA-21, miRNA-144, miRNA-208a, miRNA-34a, etc. On the other hand, a lot of studies are aimed at using miRNAs for therapeutic purposes. This review points to the need for studying the role of redox-sensitive miRNAs, to identify more effective biomarkers and develop better therapeutic targets for oxidative-stress-related heart diseases.
Collapse
Affiliation(s)
- Branislav Kura
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| | - Matus Sykora
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Jan Slezak
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.K.); (B.S.B.); (B.K.); (M.S.)
| |
Collapse
|
194
|
Savcı A, Koçpınar EF, Budak H, Çiftci M, Şişecioğlu M. The Effects of Amoxicillin, Cefazolin, and Gentamicin Antibiotics on the Antioxidant System in Mouse Heart Tissues. Protein Pept Lett 2020; 27:614-622. [PMID: 31721686 DOI: 10.2174/0929866526666191112125949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/27/2019] [Accepted: 09/18/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Free radicals lead to destruction in various organs of the organism. The improper use of antibiotics increases the formation of free radicals and causes oxidative stress. OBJECTIVE In this study, it was aimed to determine the effects of gentamicin, amoxicillin, and cefazolin antibiotics on the mouse heart. METHODS 20 male mice were divided into 4 groups (1st control, 2nd amoxicillin, 3rd cefazolin, and 4th gentamicin groups). The mice in the experimental groups were administered antibiotics intraperitoneally at a dose of 100 mg / kg for 6 days. The control group received normal saline in the same way. The gene expression levels and enzyme activities of SOD, CAT, GPx, GR, GST, and G6PD antioxidant enzymes were investigated. RESULTS GSH levels decreased in both the amoxicillin and cefazolin groups, while GR, CAT, and SOD enzyme activities increased. In the amoxicillin group, Gr, Gst, Cat, and Sod gene expression levels increased. CONCLUSION As a result, it was concluded that amoxicillin and cefazolin caused oxidative stress in the heart, however, gentamicin did not cause any effects.
Collapse
Affiliation(s)
- Ahmet Savcı
- Department of Chemistry, Faculty of Art and Science, Bingol University, Bingol, Turkey
| | - Enver Fehim Koçpınar
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Mus Alparslan University, Mus, Turkey
| | - Harun Budak
- Department of Molecular Biology and Genetics, Faculty of Science, Ataturk University, Erzurum, Turkey
| | - Mehmet Çiftci
- Department of Chemistry, Faculty of Art and Science, Bingol University, Bingol, Turkey
| | - Melda Şişecioğlu
- Department of Molecular Biology and Genetics, Faculty of Science, Ataturk University, Erzurum, Turkey
| |
Collapse
|
195
|
Kim M, Sahu A, Hwang Y, Kim GB, Nam GH, Kim IS, Chan Kwon I, Tae G. Targeted delivery of anti-inflammatory cytokine by nanocarrier reduces atherosclerosis in Apo E−/- mice. Biomaterials 2020; 226:119550. [DOI: 10.1016/j.biomaterials.2019.119550] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/26/2019] [Accepted: 10/14/2019] [Indexed: 12/19/2022]
|
196
|
TIPE1 accelerates atherogenesis by inducing endothelial dysfunction in response to oxidative stress. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165578. [PMID: 31672550 DOI: 10.1016/j.bbadis.2019.165578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/16/2019] [Accepted: 10/24/2019] [Indexed: 11/23/2022]
Abstract
Atherosclerosis is an inflammatory disease of the arterial wall, which involves endothelial cells and immune cells. Endothelial dysfunction has been considered an important step in the initiation of the disease. TIPE1 is a newly identified protein of the TIPE family, and plays a vital role in inflammation and tumorigenesis. However, its role in atherogenesis remains unclear. In this study, we demonstrated that TIPE1 promoted atherogenesis by inducing endothelial dysfunction. When human umbilical vein endothelial cells (HUVECs) were exposed to oxidative stress, the level of TIPE1 was significantly up-regulated, and the ROS generation markedly increased in TIPE1 over-expressing HUVECs. As a result, the growth of HUVECs was inhibited, and the apoptosis was enhanced. However, the cell contact ability between HUVECs and THP-1 cells were augmented due to the up-regulation of adhesion molecules such as E-selectin and ICAM-1 induced by TIPE1 overexpression. Importantly, ApoE-/- mice injected with TIPE1 recombinant lentivirus developed significantly severe atherosclerosis accompanied by hyperglycemia, hypercholesterolemia and increased white blood count. These findings indicated that excessive ROS induced by the overexpression of TIPE1 in endothelial cells accelerated the process of atherogenesis.
Collapse
|
197
|
Norton CE, Jacobsen NL, Sinkler SY, Manrique-Acevedo C, Segal SS. Female sex and Western-style diet protect mouse resistance arteries during acute oxidative stress. Am J Physiol Cell Physiol 2019; 318:C627-C639. [PMID: 31891519 DOI: 10.1152/ajpcell.00342.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A Western-style diet (WD; high in fat and carbohydrates) increases vascular oxidative stress. We hypothesized that vascular cells adapt to a WD by developing resilience to oxidative stress. Male and female C57BL/6J mice (4 wk of age) were fed a control diet (CD) or a WD for 16-20 wk. Superior epigastric arteries (SEAs; diameter, ~125 µm) were isolated and pressurized for study. Basal reactive oxygen species production was greatest in SEAs from males fed the WD. During exposure to H2O2 (200 μM, 50 min), propidium iodide staining identified nuclei of disrupted endothelial cells (ECs) and smooth muscle cells (SMCs). For mice fed the CD, death of SMCs (21%) and ECs (6%) was greater (P < 0.05) in SEAs from males than females (9% and 2%, respectively). WD consumption attenuated cell death most effectively in SEAs from males. With no difference at rest, H2O2 increased intracellular Ca2+ concentration ([Ca2+]i) to the greatest extent in SEAs from males, as shown by fura 2 fluorescence. Selective disruption of the endothelium (luminal air bubble) increased [Ca2+]i and SMC death during H2O2 exposure irrespective of sex; the WD reduced both responses most effectively in males. Nonselective transient receptor potential (TRP) channel inhibition (ruthenium red, 5 μM) attenuated the rise of [Ca2+]i, as did selective inhibition of TRP vanilloid type 4 (TRPV4) channels (HC-067047, 1 μM), which also attenuated cell death. In contrast, inhibition of voltage-gated Ca2+ channels (diltiazem, 50 μM) was without effect. Thus, for resistance arteries during acute oxidative stress: 1) ECs are more resilient than (and can protect) SMCs, 2) vessels from females are inherently more resilient than those from males, and 3) a WD increases vascular resilience by diminishing TRPV4 channel-dependent Ca2+ entry.
Collapse
Affiliation(s)
- Charles E Norton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Nicole L Jacobsen
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Shenghua Y Sinkler
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Camila Manrique-Acevedo
- Department of Medicine, University of Missouri, Columbia, Missouri.,Research Services, Harry S Truman Memorial Veterans Hospital, Columbia, Missouri.,Dalton Cardiovascular Research Center, Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
198
|
Tofas T, Draganidis D, Deli CK, Georgakouli K, Fatouros IG, Jamurtas AZ. Exercise-Induced Regulation of Redox Status in Cardiovascular Diseases: The Role of Exercise Training and Detraining. Antioxidants (Basel) 2019; 9:antiox9010013. [PMID: 31877965 PMCID: PMC7023632 DOI: 10.3390/antiox9010013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023] Open
Abstract
Although low levels of reactive oxygen species (ROS) are beneficial for the organism ensuring normal cell and vascular function, the overproduction of ROS and increased oxidative stress levels play a significant role in the onset and progression of cardiovascular diseases (CVDs). This paper aims at providing a thorough review of the available literature investigating the effects of acute and chronic exercise training and detraining on redox regulation, in the context of CVDs. An acute bout of either cardiovascular or resistance exercise training induces a transient oxidative stress and inflammatory response accompanied by reduced antioxidant capacity and enhanced oxidative damage. There is evidence showing that these responses to exercise are proportional to exercise intensity and inversely related to an individual’s physical conditioning status. However, when chronically performed, both types of exercise amplify the antioxidant defense mechanism, reduce oxidative stress and preserve redox status. On the other hand, detraining results in maladaptations within a time-frame that depends on the exercise training intensity and mode, as high-intensity training is superior to low-intensity and resistance training is superior to cardiovascular training in preserving exercise-induced adaptations during detraining periods. Collectively, these findings suggest that exercise training, either cardiovascular or resistance or even a combination of them, is a promising, safe and efficient tool in the prevention and treatment of CVDs.
Collapse
|
199
|
Abstract
The microcirculation maintains tissue homeostasis through local regulation of blood flow and oxygen delivery. Perturbations in microvascular function are characteristic of several diseases and may be early indicators of pathological changes in the cardiovascular system and in parenchymal tissue function. These changes are often mediated by various reactive oxygen species and linked to disruptions in pathways such as vasodilation or angiogenesis. This overview compiles recent advances relating to redox regulation of the microcirculation by adopting both cellular and functional perspectives. Findings from a variety of vascular beds and models are integrated to describe common effects of different reactive species on microvascular function. Gaps in understanding and areas for further research are outlined. © 2020 American Physiological Society. Compr Physiol 10:229-260, 2020.
Collapse
Affiliation(s)
- Andrew O Kadlec
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - David D Gutterman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Medicine-Division of Cardiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
200
|
Wu B, Yue H, Zhou GH, Zhu YY, Wu TH, Wen JF, Cho KW, Jin SN. Protective effects of oxymatrine on homocysteine-induced endothelial injury: Involvement of mitochondria-dependent apoptosis and Akt-eNOS-NO signaling pathways. Eur J Pharmacol 2019; 864:172717. [DOI: 10.1016/j.ejphar.2019.172717] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/26/2019] [Accepted: 10/02/2019] [Indexed: 12/19/2022]
|